WO2006071441A2 - Antibodies directed to gpnmb and uses thereof - Google Patents

Antibodies directed to gpnmb and uses thereof Download PDF

Info

Publication number
WO2006071441A2
WO2006071441A2 PCT/US2005/043482 US2005043482W WO2006071441A2 WO 2006071441 A2 WO2006071441 A2 WO 2006071441A2 US 2005043482 W US2005043482 W US 2005043482W WO 2006071441 A2 WO2006071441 A2 WO 2006071441A2
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
gpnmb
seq
antibodies
cells
Prior art date
Application number
PCT/US2005/043482
Other languages
French (fr)
Other versions
WO2006071441A3 (en
Inventor
Feng Xiao
Xiao-Chi Jia
Meina Liang
Orit Foord
Scott Klakamp
Kam Fai Tse
Vincent A. Pollack
Luca Rastelli
John Herrmann
Henri Lichensten
Michael E. Jeffers
William J. Larochelle
Gulshan Ara
Peter Mezes
Andrei Chapoval
Cyrus Karkaria
Michael Torgov
Juan Davagnino
Original Assignee
Curagen Corporation
Amgen Fremont, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to EP05852645A priority Critical patent/EP1827492B1/en
Priority to DE602005022928T priority patent/DE602005022928D1/en
Priority to AT05852645T priority patent/ATE476994T1/en
Priority to DK05852645.0T priority patent/DK1827492T3/en
Priority to CA2589374A priority patent/CA2589374C/en
Priority to AU2005322410A priority patent/AU2005322410B2/en
Application filed by Curagen Corporation, Amgen Fremont, Inc. filed Critical Curagen Corporation
Priority to JP2007543622A priority patent/JP2008521411A/en
Publication of WO2006071441A2 publication Critical patent/WO2006071441A2/en
Publication of WO2006071441A3 publication Critical patent/WO2006071441A3/en
Priority to US13/355,366 priority patent/US8846873B2/en
Priority to US14/454,179 priority patent/US20150044212A1/en
Priority to US15/053,204 priority patent/US20160311919A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3053Skin, nerves, brain
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6849Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a receptor, a cell surface antigen or a cell surface determinant
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6851Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2809Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against the T-cell receptor (TcR)-CD3 complex
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3061Blood cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/545Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • C07K2317/732Antibody-dependent cellular cytotoxicity [ADCC]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/77Internalization into the cell
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/94Stability, e.g. half-life, pH, temperature or enzyme-resistance

Definitions

  • the present invention relates to antibodies with specificity to GPNMB, and uses of such antibodies.
  • the present invention provides fully human monoclonal antibodies that specifically bind to GPNMB, and uses thereof.
  • Nucleotide sequences encoding, and amino acid sequences comprising, heavy and light chain immunoglobulin molecules, particularly sequences corresponding to contiguous heavy and light chain sequences spanning the framework regions and/or complementarity determining regions (CDRs) are provided.
  • the present invention also provides immunoconjugates comprising anti-GPNMB antibodies and methods of using such immunoconjugates.
  • the present invention further provides bi-specific antibodies comprising an anti-GPNMB antibody component and an anti-CD3 component, and methods of using such bispecific antibodies.
  • GPNMB A putative transmembrane glycoprotein called "ninV (Ace. No. X76534 EMBL) ,
  • GPNMB GPNMB ' referred to herein as GPNMB
  • Weterman et al. (Int J Cancer 60:73-81, 1995) as differentially expressed in low-metastatic human melanoma cancer cell lines and xenografts, compared to a more aggressive melanoma cell line.
  • GPNMB shares 33% identity with the precursor of pMel 17 melanocyte-specific protein (Kwon et al , 1991 , PNAS 88:9228-9232).
  • GPNMB is 71 % homologous to a dendiitic cell- associated transmembrane protein, DC-HIL (Shikano et al, 2001 Biol. Chem. 276:8125- 8134).
  • GPNMB is also known as the hematopoietic growth factor inducible neurokinin- 1 protein HGFIN (Bandari et al, Reg. Peptides 111 :169-178) and the bone-related gene osteoactivin (Owen et al. Crit Rev Eukaryot Gene Expr 2003, 13(2-4):205-220) It was also reported that nmb could reduce the metastatic potential of a highly metastatic mnb-negative melanoma cell line (Weterman, 1995). GPNMB was considered a candidate glioblastoma tumor marker after public database mining and expression profiling (Loging et al, 2000, Genome Research 10: 1393-1402).
  • NCBI SAGE data also shows overexpression of this gene in stomach and pancreatic carcinoma.
  • the mouse ortholog has been shown to be highly upregulated in a neural stem cell line NSC, derived from the TSC2 knockout model for Tuberous Sclerosis Complex Syndrome
  • Antibodies also known as immunoglobulins, are typically tetrameric glycosylated proteins composed of two light (L) chains (about 25 kDa) and two heavy (H) chains (about 50-70 kDa).
  • the amino-terminal portion of each chain includes a variable domain of about 100 to 110 or more amino acids primarily responsible for antigen recognition.
  • the carboxy- terminal portion of the L and H chain has one and three or four constant domains, respectively that are primarily responsible for effector function.
  • H chains are classified as mu, delta, gamma, alpha, or epsilon based upon the constant domain amino acid sequence, defining the antibody's isotype as IgM, IgD, IgG, IgA, and IgE, respectively.
  • Isotypes may be further divided into subclasses e.g. IgGi, IgG 2 , IgG 3 , IgG 4 .
  • Immunoglobulins can be produced naturally in vivo by B lymphocytes. Each clone of B cells produces antibody with an antigen receptor having a unique prospective antigen binding structure. The repertoire of antigen receptors, approximately 10 7 possibilities, exists in vivo prior to antigen stimulation. This diversity is produced by somatic recombination, i.e., the joining of different antibody gene segments. Immunoglobulin H chain, kappa L chain and lambda L chain are encoded by three separate genetic loci and each locus has multiple copies of at least 3 types of gene segments encoding variable (V), constant (C) and joining (J) regions, the heavy chain gene also includes a diversity (D) region.
  • V variable
  • C constant
  • J joining
  • V, C and J regions and D for the heavy chain
  • the joining of V, C and J regions can result in the loss or addition of residues at the junctions.
  • the L and H chain V region of human antibodies consists of relatively conserved framework regions (FR) that form a scaffold for three hypervariable regions also known as complementary determining regions (CDR). From the amino terminus of either the heavy or light chain, the V domain is made up of FR and CDR regions in the following order: FRl -CDRl -FR2-CDR2-FR3.
  • CDR3-FR4 Joining of the V domain with a D (heavy chain only) and J domain adds CDR3-FR4.
  • the CDRs are generally responsible for antigen binding.
  • the specificity of monoclonal antibodies have made them attractive agents for targeting cancer in vivo with the hopes of eradicating disease while sparing normal tissue.
  • the approach, which initially utilized mouse monoclonal antibodies has encountered limitations to potential effectiveness such as immunogenicity; inefficient effector functions and short half-life in vivo.
  • chimeric antibodies which sought to utilize the antigen binding variable domains of mouse monoclonal antibodies combined with the constant regions of human antibodies (Boulianne, et al 1984 Nature 312:643-646; Morrison et al, 1984 PNAS USA 81:6851-6855); humanized antibodies which grafted antigen binding complementary determining regions (CDRs) from mouse antibodies to human immunoglobulin (Jones, et al, 1986 Nature 321: 522-525; Riechmann, et al, 1988 Nature 332:323-327; Verhoeyen, et al, 1988 Science 239:1534-1536; Vauglum, et al, 1998 Nature Biotechnol.
  • CDRs complementary determining regions
  • transgenic animals having human immunoglobulin genes and nonfunctional endogenous genes have been developed for immunization and production of fully human monoclonal antibodies (Fislrwild, et al, 1996 Nature Biotechnol 14:845-851; Mendez, et al, 1997 Nature Genet. 15:146-156; Nicholson, et al, 1999 J. Immunol 163, 6898-6906).
  • Single Chain Antibodies Single Chain Antibodies: Single chain Fv antibodies (scFvs) were first described in the late 1980 1 S (Bird et al, Science 242:423-426 (1988); Huston et al, Proc. Natl. Acad. Sci. USA 85:5879-5883 (1988)).
  • a polypeptide linker typically ranging in length from 5 to 27 amino acid residues, is used to join the C-terminus of the variable light chain domain (V L ) to the N-terminus of the variable heavy chain domain (VH).
  • V L variable light chain domain
  • VH variable heavy chain domain
  • the linker joins the C-terminus of the VH to the N-terminus of the V L -
  • fo ⁇ nats V L -VH and V H - VL
  • the most common linker used in the literature is the (Gly 4 Ser) 3 15 amino acid linker, however there are several other linkers that have been utilized, including a 25 amino acid linker called 205C (Pantoliano et al, Biochemistry 30:10117-10125 (1991)).
  • Bispecific Antibodies An area of mAb research where considerable progress has been made is in the development of bispecific antibodies (biAbs). There are distinct advantages to developing therapeutic antibody molecules with dual specificity. For example, biAbs can serve as mediators to target immune effector cells such as CTLs to unwanted cells (Baeuerle et al., Curr. Opin. MoI. Ther.
  • bispecific antibodies directed against Fc gamma receptors CD 16, CD64, and CD89 were significantly more effective in vitro than conventional IgG antibodies (Peipp and Valerius, Biochem. Soc. Trans. 30:507-511 (2002)).
  • Fc gamma receptors CD 16, CD64, and CD89 were significantly more effective in vitro than conventional IgG antibodies.
  • One of the challenges in developing biAbs as viable therapeutics has been producing large enough quantities of a stable moiety for clinical applications.
  • Another challenge has been in determining the right combination of validated targets and the underlying biology that would lead to a therapeutic product. For recent reviews on the difficulties experienced with biAbs, see (Kontermann, Acta Pharmacol Sin 26:1-9 (2005); Peipp and Valerius, Soc. Trans. 30:507-511 (2002)).
  • Bi-scFv Bispecific Single Chain Antibodies
  • Bi-scFvs are typically comprised of 4 variable domains, 2 heavy (V H ) and 2 light (V L ), which are derived from 2 different antibodies.
  • V H variable domains
  • V L variable domains
  • the 4 domains are linked together with 3 short linkers, ranging in length from 5-27 amino acids.
  • the biological activity of this type of antibody depends on several features concerning the construction of the molecule.
  • both the linker sequences between the antibody V domains and the order of the 4 antibody V domains themselves can vary, as well as the expression system that is used; all of which can greatly affect the solubility and biological activity of the various resulting products (Kipriyanov et al, J. MoI. Biol. 330:99-111 (2003); Le Gall et al, Protein Eng. Des. SeI. 17:357-366 (2004); Pavlinkova et al, Clin Cancer Res. 5:2613-1619 (1999)).
  • Cytotoxic T lymphocytes Under normal circumstances, T cells are activated when the CD3/T cell receptor (CD3/TCR) complex binds to a relevant MHC molecule associated with a specific Ag peptide. Engagement of CD3/TCR with MHC results in intracellular signals necessary to trigger an immune response against a pathogen or tumor. Similar signals that cause T cell activation can also be achieved by antibodies that can bind certain structures of the CD3/TCR complex.
  • biAbs recognizing both the TCR/CD3 complex and tumor associated antigen (TAA) can trigger the activation program in CTLs in the presence of target cells (Chapoval et al, J. Immunol 155:1296-1303 (1995)).
  • Recombinant technologies are being utilized to enable further improvements upon antibody molecules with the goal of enhancing in vivo efficacy.
  • Such technologies provide, for example, for optimizing molecular size, affinity, pharmacokinetics, toxicity, specificity, valency, effector functions, direct and indirect arming, combination therapy, and various prodrug approaches.
  • the current invention provides human monoclonal antibodies that specifically bind GPNMB as well as variants, derivatives and antigen binding fragments of such antibodies.
  • the invention provides preferred somatic recombinations of human antibody gene segments to provide specificity for GPNMB and genetically engineered anti -GPNMB antibody variants and derivatives that originate from these gene segments.
  • the current invention provides multiple affinity matured human antibodies with binding specificity for GPNMB.
  • the present invention provides an antibody, or binding fragment thereof, that binds to GPNMB, wherein said antibody, or binding fragment thereof, neutralizes a GPNMB-induced activity, and wherein said antibody, or binding fragment thereof, cross-reacts with a fully human anti-GPNMB antibody selected from the group consisting of Mab 1.2.1, Mab 1.10.1, and Mab2.22.1 or an antibody in the same antigen- binding bin as fully human anti-GPNMB antibody Mabl .2.1 , Mabl .10.1 , or Mab2.22.1.
  • a fully human anti-GPNMB antibody selected from the group consisting of Mab 1.2.1, Mab 1.10.1, and Mab2.22.1 or an antibody in the same antigen- binding bin as fully human anti-GPNMB antibody Mabl .2.1 , Mabl .10.1 , or Mab2.22.1.
  • the present invention provides an antibody, or binding fragment thereof, that binds to GPNMB, wherein said antibody, or binding fragment thereof, neutralizes a GPNMB-induced activity, and wherein said antibody, or binding fragment thereof, cross-reacts with a fully human anti-GPNMB antibody selected from the group consisting of Mab2.3.1 and Mabl.15.1 or an antibody in the same antigen-binding bin as fully human anti-GPNMB antibody Mab2.3.1 or Mabl.15.1.
  • the present invention provides an antibody, or binding fragment thereof, that binds to GPNMB, wherein said antibody, or binding fragment thereof, neutralizes a GPNMB-induced activity, and wherein said antibody, or binding fragment thereof, cross-reacts with fully human anti-GPNMB antibody Mab2.10.1 or an antibody in the same antigen-binding bin as fully human anti-GPNMB antibody Mab2.10.1.
  • the present invention provides naked IgGl anti-GPNMB antibodies that have cytotoxic effect to cells overexpressing GPNMB.
  • the present invention provides methods of treating or preventing diseases associated with overexpression of GPNMB comprising administering to a subject in need thereof a composition comprising a naked IgGl anti-GPNMB antibody and an immunomodulator (such as, but not limited to, interferons and cytokines).
  • the present invention provides immunoconjugates that comprise an anti-GPNMB antibody or a fragment thereof, and a cytotoxic agent.
  • the cytotoxic agent is auristatin E (dolastatin-10) or a derivative thereof. Methods of using such immunoconjugated are also provided.
  • the present invention provides bispecific antibodies comprising an anti-GPNMB component and an anti-CD3 antibody component, which enable the cytotoxic killing of target tumor cells by T cells.
  • the present invention provides single chain Fv antibody conjugated to a cytotoxic agent.
  • the cytotoxic agent is auristatin E (dolastatin-10) or a derivative thereof. Methods of using such bispecific antibodies and conjugated single chain Fv antibodies are also provided. Amino acid sequences for anti-GPNMB human monoclonal antibodies of the invention and nucleic acid sequences encoding them are provided.
  • compositions comprising human anti-GPNMB antibodies, including therapeutic compositions comprising same, and methods of use are provided.
  • therapeutic immunoconjugates comprising anti-GPNMB antibodies and a cytotoxic or cytostatic agent for treating GPNMB expressing cancers and other GPNMB related disorders are provided. Dosage regimens are also provided.
  • Figure 1 Tumor growth inhibition and complete regression of SK-MEL-2 xenografts in athymic mice after treatment with 2.50 to 20 mg/kg i.v. every 4 days for 4 treatments.
  • the responses of tumor-bearing animals to reference drugs such as vinblastine (1.7 mg/kg i.v. q4d X4) and paclitaxel (24 mg/kg i.v. q2d X4) are also shown.
  • Control groups are treated with either phosphate-buffered saline (PBS) or physiological saline.
  • PBS phosphate-buffered saline
  • Figure 2 Indirect immunotoxin killing of UACC-62 melanoma cells by anti- GPNMB antibodies
  • Figure 3 Inhibition of colony formation of UACC-62 cells incubated with Auristatin E (AE) conjugated anti-GPNMB antibodies.
  • AE Auristatin E
  • Figure 4 Tumor growth inhibition and complete regression of SK-MEL-2 xenografts in athymic mice after treatment with CROl 1-vcMMAE 5.0 mg/kg i.v. every 4 days for 4 treatments.
  • CROl 1-vcMMAE 5.0 mg/kg i.v. every 4 days for 4 treatments.
  • the lack of responses of tumor-bearing animals to unconjugated CROl 1 or to free monomethylauristatin E demonstrate that the intact immunoconjugate is essential for anti-tumor effects.
  • FIG. 5 Tumor size reduction and complete regression of SK-MEL-2 xenografts in athymic mice after treatment with 1.25 to 20 mg/kg i.v. every 4 days for 4 treatments.
  • the responses of tumor-bearing animals to reference drugs such as Vinblastine (1.7 mg/kg i.v. q4d X4) and paclitaxel (24 mg/kg i.v. q2d X4) are also shown.
  • Control groups are treated with either phosphate-buffered saline (PBS) or physiological saline.
  • PBS phosphate-buffered saline
  • FIG. 6 The serum concentration-time profile of the antibody of CROl 1-vcMMAE after intravenous administration of 1 and 10 mg/kg in athymic mice. Detection was achieved with a sandwich ELISA assay, which employed the CROl 1 antigen (CG56972,
  • GPNMB horseradish peroxidase-conjugated anti-human globulin. Results shown are the serum concentrations expressed as ⁇ g/mL (left x-axis) and micromolarmolar concentration (right X-axis).
  • Figure 7 Aggregate responses, expressed as percent cures, were recorded for test animals treated with 5 different, graduated dosing intervals (i.e., 0, 1, 4, 8, and 16 days between treatments). The slope of the line is not significantly different from 0 (p ⁇ 0.2904).
  • FIG. 9 Effects of ectopic expression of GPNMB or sensitivity to CROIl- vcMMAE.
  • HEK293 cells are transfected with empty vector (vector) or GPNMB-containing plasmid (GPNMB) as described in Materials and Methods.
  • A. Cell lysates are prepared from the transfected HEK293 cells and the expression of GPNMB (upper panel) or actin (lower panel) is determined by immunoblotting. Lane 1: Empty vector transfectants. Lane 2: GPNMB transfectants.
  • B Flow cytometry analysis of GPNMB expression on empty vector or GPNMB transfected cells.
  • Cells are treated with various concentrations of CRO 11 -vcMMAE (diamonds: vector or circles: GPNMB) or IgG2-vcMMAE (triangles: vector or squares: GPNMB) for 96 hours. After a clonogenic assay, the surviving fraction is normalized to the untreated control and expressed as a percentage of the control using GraphPad Prism graphing software. Each treatment is performed in triplicate. A representative graph from two independent experiments is shown.
  • Figure 10 Effect of GPNMB siRNA on endogenous GPNMB expression and sensitivity to CRO 11 -vcMMAE.
  • SK-Mel-2 cells are transfected with 50 nJVI of control siRNA or siRNA tai'geting GPNMB.
  • A. Cell Iy sates are prepared from the transfected SK- Mel-2 cells 2 and 4 days post-transfection and the expression of GPNMB (upper panel) or actin (lower panel) is determined by immunoblotting. Lane 1 : Mock (oligofectamine) transfection. Lane 2: Control siRNA transfection. Lane 3: GPNMB siRNA transfection.
  • B Flow cytometry analysis of GPNMB expression 2 and 4 days after transfection.
  • SK-Mel-2 cells are transfected with mock, control siRNA or GPNMB siRNA as indicted in the Materials and Methods.
  • C. CROl 1 -vcMMAE in vitro growth inhibition of mock (diamonds), control siRNA (circles) or GPNMB siRNA (triangles) transfected SK-Mel-2 cells is determined by a clonogenic assay as described in Materials and Methods. The surviving fraction is normalized to the untreated control and expressed as a percentage of control using GraphPad Prism graphing software. Each treatment is performed in triplicate. A representative experiment from two independent studies is shown.
  • Figure 11 FACS analysis of SK-MEL-2 with isotype control, hybridoma IgG2
  • Figure 12 (A) PBMC and mAb (IgGl) mediated ADCC of SK-MEL-2 cells. ADCC effector functions are measured as described above at 2, 5 and 10 ⁇ g/200 ⁇ l using targeteffector ratios of 10, 30, 60 and 100 as indicated. (B) PBMC and mAb (IgG2) do not cause ADCC to SK-MEL-2 cells. ADCC effector functions are measured as described above at 0, 2, 5 and 10 ⁇ g/200 ⁇ l using target: effector ratios of 10, 30, 60 and 100 as indicated.
  • Figure 13 Expression of CG56972 in human cancer cell lines and tissues.
  • RTQ PCR analysis of A) human brain cancer cell lines or (B) human brain cancer glioma and medulloblastoma biopsies.
  • C Microarray analysis of CG56972 expression in human brain cancer and oligodendroglioma tissues. Tissues or cell lines are harvested, mRNA prepared and RTQ PCR or CuraChip analysis performed as described in Materials and Methods.
  • Figure 14 FACS analysis of cell surface binding of CROl 1 mAb to CG56972.
  • SK- MEL-2, XF-498, U-118-MG, SNB-78, SF-539 and SF-268 cells are labeled with a saturating concentration (10 ⁇ g/mL) of CROl 1 mAb or control IgG2.
  • Bound mAb is detected by flow cytometry with PE-conjugated goat-anti-human secondary antibody as described in Materials and Methods.
  • GM Geometric mean.
  • the SF-268 cell line is CG56972 transcript negative and used as a negative control.
  • Figure 15 Immunoblot analysis of CG5672 expression in human brain cancer cell lines. Cell lysates are resolved on Tris-glycine gels and transferred to membranes. Immunoblot analysis is carried out with a polyclonal antibody to CG56972 followed by enhanced chemiluminescence detection as described in Materials and Methods. Arrowheads indicate the relative mobility of the plOO and 120 CG56972 species.
  • the SF-268 cell line is CG56972 transcript negative and used as a negative control.
  • FIG. 16 CROl 1-vcMMAE in vitro growth inhibition of astocytoma/glioblastoma cell growth.
  • XF-498, SNB-78, U-118-MG, SF-539, LOXIMVI and SF-268 cells are incubated with the indicated concentration of CROl 1-vcMMAE.
  • Cells are also incubated with control PKl 6.3 mAb (data shown in Table I) as described in the Materials and Methods. Cell growth was determined by clonogenic assay. The surviving colonies are counted and plotted using GraphPad Prism graphing software. The experiment is performed in triplicate wells and repeated twice. vA representative experiment is shown. IC50s for cell killing is presented in ng/mL concentrations.
  • the LOXIMVI and SF-268 cell lines are CG56972 transcript negative and used as negative controls.
  • FIG. 17 Development of CROI l Engineered Antibodies.
  • V antibody variable
  • VL and VH light and heavy chain variable domains
  • the middle linker joining the 2 individual scFv components together may play a key role in determining the resulting activity of each of the scFv components, including the effective cytolytic activity provided by the cytotoxic T cells engaged by the anti-CD3 scFv component of the bi-scFv.
  • Figure 18 A.
  • Figure 19 Flow cytometry analysis of binding of CROl 1 scFv and CROl 1 x anti- CD3 (L4-L2-L4 linker set) bi-scFv products to native GPNMB protein expressed on the cell surface of target cells.
  • Human T cells are used as a source of CD3, while SK-Mel-5 cells are used as a source of GPNMB.
  • FIG. 20 Cytotoxicity analysis showed that purified CROl 1 x anti-CD3 (L4-L2-L4 linker set) bi-scFv, but not CROl 1 scFv, causes killing of GPNMB positive SK-Mel-5 tumor cells by T lymphocytes.
  • Figure 21 The chemical structure of Maleimidocoaproyl-Valine-Citrullin-
  • Figure 22 Disulfides on CROl 1 antibody are gently reduced in the presence of TCEP to generate ⁇ 4 thiols per Ab. vcMMAE is then added to antibody solution. Nucleophilic attack of thiolates on maleimide-groups results in a stable thioester linkage. The resulting conjugate is purified from the mixture.
  • Figure 23 Reaction of vcMMAE with NAcCys at pH 7.0 and pH 9.0 in the presence or absence of TCEP.
  • IA VCMMAE converts fully into NAcCys-adduct following a incubation in phosphate pH 7 buffer.
  • B-E Appearance of a side product in a course of incubation of vcMMAE in borate buffer.
  • F-I Appearance of side products in borate pH 9 and in the presence of TCEP.
  • Figure 24 LCMS identification of the side product with retention time of 9.2 min not capable of reaction with cystein and therefore, not capable of conjugation to CROl 1.
  • Figure 25 Kinetics of the formation of NAcCys-vcMMAE and of the side product (succinimidyl-vcMMAE) following incubation in borate pH 9.0 buffer in the presence or absence of TCEP.
  • antibody refers to an immunoglobulin or a fragment or a derivative thereof, and encompasses any polypeptide comprising an antigen-binding site, regardless whether it is produced in vitro or in vivo.
  • the term includes, but is not limited to, polyclonal, monoclonal, monospecific, polyspecific, non-specific, humanized, single-chain, chimeric, synthetic, recombinant, hybrid, mutated, engineered, and grafted antibodies.
  • antibody also includes antibody fragments such as Fab, F(ab') 2 , Fv, scFv, bi-scFv, bi-Ab, Fd, dAb, and other antibody fragments that retain antigen-binding function, i.e., the ability to bind GPNMB specifically. Typically, such fragments would comprise an antigen-binding domain.
  • the terms "antigen-binding domain,” “antigen-binding fragment,” and “binding fragment” refer to a part of an antibody molecule that comprises amino acids responsible for the specific binding between the antibody and the antigen. In instances, where an antigen is large, the antigen-binding domain may only bind to a part of the antigen. A portion of the antigen molecule that is responsible for specific interactions with the antigen-binding domain is referred to as “epitope" or "antigenic determinant.”
  • An antigen-binding domain typically comprises an antibody light chain variable region (V L ) and an antibody heavy chain variable region (V H ), however, it does not necessarily have to comprise both.
  • V L antibody light chain variable region
  • V H antibody heavy chain variable region
  • a so-called Fd antibody fragment consists only of a VH domain, but still retains some antigen-binding function of the intact antibody.
  • the term "repertoire” refers to a genetically diverse collection of nucleotides derived wholly or partially from sequences that encode expressed immunoglobulins .-The sequences are generated by in vivo rearrangement of, e.g., V, D, and J segments for H chains and, e.g., V and J segment for L chains. Alternatively, the sequences may be generated from a cell line by in vitro stimulation, in response to which the rearrangement occurs. Alternatively, part or all of the sequences may be obtained by combining, e.g.
  • binding refers to two molecules forming a complex that is relatively stable under physiologic conditions. Specific binding is characterized by a high affinity and a low to moderate capacity as distinguished from nonspecific binding which usually has a low affinity with a moderate to high capacity. Typically, binding is considered specific when the affinity constant K A is higher than 10 6 M " 1 , or more preferably higher than 10 8 M "1 . If necessary, non-specific binding can be reduced without substantially affecting specific binding by varying the binding conditions.
  • the appropriate binding conditions such as concentration of antibodies, ionic strength of the solution, temperature, time allowed for binding, concentration of a blocking agent (e.g., serum albumin, milk casein), etc., may be optimized by a skilled artisan using routine techniques.
  • the term "substantially as set out” refers that the relevant CDR, V H , or V L domain of the invention will be either identical to or have only insubstantial differences in the specified regions (e.g., a CDR), the sequence of which is set out. Insubstantial differences include minor amino acid changes, such as substitutions of 1 or 2 out of any 5 amino acids in the sequence of a specified region.
  • CROl 1 refers to a fully human monoclonal antibody that specifically binds to GPNMB. In some embodiments, CROl 1 refers to those antibodies that are identified in Tables 2A-2D of the present application. In some embodiments, CROl 1 refers to Mab 1.15.1 as described in the instant invention.
  • GPNMB transmembrane glycoprotein that has an amino acid sequence as set forth in SEQ ID NO: 289, an analog, derivative or a fragment thereof, or a fusion protein comprising GPNMB, an analog, derivative or a fragment thereof.
  • GPNMB refers to the mature, processed form of GPNMB.
  • GPNMB refers to the extracellular domain of GPNMB.
  • GPNMB activity refers to one or more activities associated with GPNMB.
  • To “modulate” GPNMB activity is to alter the baseline results observed with, and that can be attributed to GPNMB.
  • To "neutralize” GPNMB is to cancel one or more effects, e.g. activity observed with, and that can be attributed to GPNMB.
  • isolated refers to a molecule that is substantially • free of its natural environment.
  • an isolated protein is substantially free of cellular material or other proteins from the cell or tissue source from which it is derived.
  • isolated also refers to preparations where the isolated protein is sufficiently pure to be administered as a pharmaceutical composition, or at least 70-80% (w/w) pure, more preferably, at least 80-90% (w/w) pure, even more preferably, 90-95% pure; and, most preferably, at least 95%, 96%, 97%, 98%, 99%, or 100% (w/w) pure.
  • the term “inhibit” or “inhibition of” refers to reducing by a measurable amount, or to prevent entirely.
  • the term “Cytotoxic effect” in reference to the effect of an agent on a cell means killing of the cell.
  • “Cytostatic effect'” refers to an inhibition of cell proliferation.
  • a “cytotoxic agent” refers an agent that has a cytotoxic or cytostatic effect on a cell, thereby depleting or inhibiting the growth of, respectively, cells within a cell population.
  • the te ⁇ ns "prevent,” “preventing,” and “prevention” refer to the inhibition of the development or onset of a disorder associated with aberrant expression and/or activity of GPNMB (e.g., cancer) or the prevention of the recurrence, onset, or development of one or more symptoms of a disorder associated with aberrant expression and/or activity of GPNMB (e.g., cancer) in a subject resulting from the administration of a therapy or the administration of a combination of therapies.
  • the term "effective amount” refers to a dosage or amount that is sufficient to reduce the activity of GPNMB to result in amelioration of symptoms in a patient or to achieve a desired biological outcome.
  • prophylactically effective amount refers to the amount of a therapy which is sufficient to result in the prevention of the development, recurrence, or onset of a disorder associated with aberrant expression and/or activity of GPNMB (e.g., cancer) or one or more symptoms thereof, or to enhance or improve the prophylactic effect(s) of another therapy.
  • GPNMB e.g., cancer
  • a “protocol” includes dosing schedules and dosing regimens.
  • the protocols herein are methods of use and include prophylactic and therapeutic protocols.
  • the terms “subject” and “patient” are used interchangeably.
  • the terms “subject” and “subjects” refer to an animal, preferably a mammal including a non-primate (e.g., a cow, pig, horse, cat, dog, rat, and mouse) and a primate (e.g., a monkey, such as a cynomolgous monkey, chimpanzee, and a human), and more preferably a human.
  • the terms “therapeutic agent” and “therapeutic agents” refer to an agent that can be used in the prevention, treatment, management, or amelioration of a disorder associated with aberrant expression and/or activity of GPNMB (e.g., cancer) or one or more symptoms thereof.
  • the term “therapeutic agent” refers to an antibody that immunospecifically binds to GPNMB.
  • the term “therapeutic agent” refers an agent other than an antibody that immunospecifieally binds to GPNMB.
  • the terms “therapies” and “therapy” can refer to any protocol(s), method(s), and/or agent(s) that can be used in the prevention, treatment, management, or amelioration of a disorder associated with aberrant expression and/or activity of GPNMB (e.g., cancer) or one or more symptoms thereof.
  • the terms “therapies” and “therapy” refer to anti-cancer therapy, biological therapy, supportive therapy, and/or other therapies useful in treatment, management, prevention, or amelioration of cancer or one or more symptoms thereof known to one of skill in the art such as medical personnel.
  • the terms “treat,” “treatment,” and “treating” refer to the eradication, removal, modification, or control of primary, regional, or metastatic cancer tissue, or the reduction or amelioration of the progression, severity, and/or duration of a disorder associated with aberrant expression and/or activity of GPNMB or amelioration of one or more symptoms thereof resulting from the administration of one or more therapies.
  • such terms in the context of cancer refer to a reduction in the growth of cancerous cells, a decrease in number of cancerous cells and/or a reduction in the growth, formation and/or volume of a tumor.
  • such terms refer to the minimizing or delay of the spread of cancer resulting from the administration of one or more therapies to a subject with such a disease.
  • Treatment can include, for example, a decrease in the severity of a sypmtopm, the number of symptoms, or frequency of relapse.
  • scientific and technical terms used in connection with the invention described herein shall have the meanings that are commonly understood by those of ordinary skill in the art. Further, unless otherwise required by context, singular terms shall include pluralities and plural terms shall include the singular.
  • nomenclatures utilized in connection with, and techniques of, cell and tissue culture, molecular biology, and protein and oligo- or polynucleotide chemistry and hybridization described herein are those well known and commonly used in the art.
  • Standard techniques are used for recombinant DNA, oligonucleotide synthesis, and tissue culture and transformation (e.g., electroporation, lipofection). Enzymatic reactions and purification techniques are performed according to manufacturer's specifications or as commonly accomplished in the art or as described herein. The foregoing techniques and procedures are generally performed according to conventional methods well known in the art and as described in various general and more specific references that are cited and discussed throughout the present specification. (See e.g., Sambrook et a Molecular Cloning: A Laboratory Manual, 2d ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, K Y. 1989).
  • the nomenclatures utilized in connection with, and the laboratory procedures and techniques of, analytical chemistry, synthetic organic chemistry, and medicinal and pharmaceutical chemistry described herein are those well known and commonly used in the art. Standard techniques are used for chemical syntheses, chemical analyses, pharmaceutical preparation, formulation, and delivery, and treatment of patients.
  • the current invention provides germline human antibody heavy chain V, D, J combinations and light chain V, J combinations including nucleotide and amino acid sequence of the V H and VL domain FR and CDR regions with specificity for GPNMB.
  • the current invention provides the nucleotide and amino acid sequence of such affinity matured V domain FR and CDR regions having specificity to GPNMB.
  • Fab type antibody fragments containing the antigen binding portion of the antibody molecule may consist of the L chain covalently linked by a disulfide bond to a portion of the H chain which has the V domain and first constant domain.
  • Single chain Fv antibody fragment (scFv) has the H variable domain linked to the L variable domain by a polypeptide linker.
  • the invention provides antibody fragments such as Fab and scFv molecules having sequences derived from germline or affinity matured V domains of antibodies binding specifically to GPNMB.
  • a bispecific or bifunctional antibody is an artificial hybrid antibody having two different heavy/light chain pairs and two different binding sites.
  • Bispecific antibodies can be produced by a variety of methods including fusion of hybridomas or linking of Fab * fragments (see, e.g., Songsivilai & Lachmann, 1990 Clin. Exp. Immunol. 79: 315-321; Kostelny et ah, 1992 J. Immunol. 148:1547-1553).
  • Bispecific antibodies do not exist in the form of fragments having a single binding site (e.g., Fab, Fab ⁇ and Fv).
  • bifunctional or bispecific antibodies are contemplated and encompassed by the invention.
  • a bispecific single chain antibody with specificity to GPNMB and to the CD3 antigen on cytotoxic T lymphocytes can be used to direct these T cells to tumor cells expressing GPNMB and cause apoptosis and eradication of the tumor.
  • Bispecific scFv constructs for this purpose are described herein.
  • the scFv components specific for GPNMB can be derived from anti-GPNMB antibodies described herein.
  • the anti-GPNMB antibody components disclosed herein can be used to generate a biologically active scFv directed against GPNMB.
  • the anti-CD3 scFv component of the therapeutic bispecific scFv was derived from a sequence deposited in Genbank (accession number CAE85148).
  • Alternative antibodies known to target CD3 or other T cell antigens may similarly be effective in treating malignancies when coupled with anti-GPNMB, whether on a single-chain backbone or a full IgG.
  • GPNMB binding human antibodies may include H or L constant domains including L kappa or lambda constant regions, or any isotype H constant domain.
  • a human antibody with binding specificity to GPNMB contains germline sequences such as the heavy chain V regions: VH 1-2 (SEQ ID NO: 308), VH2-5 (SEQ ID NO: 360), VH3-11 (SEQ ID NO: 361), VH3-21 (SEQ ID NO: 362), VH3-30 (SEQ ID NO:363), VH3-33 (SEQ ID NO: 364), VH4-31 (SEQ ID NO: 365), VH4-59 (SEQ ID NO:366) or VH5-51 (SEQ ID NO:367); the heavy chain D region: Dl-20 (amino acid sequences translated by SEQ ID NO: 375), Dl -26 (amino acid sequences translated by SEQ ID NO:376), D3-10 (amino acid sequences translated by SEQ ID NO:377),
  • human antibodies with binding specificity to GPNMB are combined germline regions as shown in Table 1.
  • the isolated antibody has a heavy chain variable region polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NOs:2, 20, 38, 56, 74, 92, 110, 128, 146, 164, 182, 200, 218, 236, 253, 256, 260, 265, 270, 274, 277, 281 and 285.
  • amino acid sequences can be encoded by nucleotide sequences selected from the group consisting of SEQ ID NOs: 1, 19, 37, 55, 73, 91, 109, 127, 145, 163, 181, 199, 217 and 235.
  • the invention provides an isolated antibody that specifically binds to GPNMB and has a light chain variable region polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 11, 29, 47, 65, 83, 101, 119, 137, 155, 173, 191, 209, 227 and 245.
  • amino acid sequences can be encoded by nucleotide sequences selected from the group consisting of SEQ ID NOs: 10, 28, 46, 64, 82, 100, 118, 136, 154, 172, 190, 208, 226 and 244.
  • the invention provides an isolated antibody that specifically binds to GPNMB and has a heavy chain polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 2, 20, 38, 56, 74, 92, 110, 128, 146, 164, 182, 200, 218, 236, 253, 256, 260, 265, 270, 274, 277, 281 and 285 and has a light chain polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 11, 29, 47, 65, 83, 101, 119, 137, 155, 173, 191, 209, 227 and 245.
  • anti-GPNMB antibodies comprise at least one CDR of any of the H or L CDR polypeptide sequences SEQ ID NOs: 4, 6, 8, 13, 15, 17, 22, 24, 26, 31, 33, 35, 40, 42, 44, 49, 51, 53, 58, 60, 62, 67, 69, 71, 76, 78, 80, 85, 87, 89, 94, 96, 98, 103, 105, 107, 112, 114, 116, 121, 123, 125, 130, 132, 134, 139, 141, 143, 148, 150, 152, 157, 159, 161, 166, 168, 170, 175, 177, 179, 184, 186, 188, 193, 195, 197, 202, 204, 206, 211, 213, 215, 220, 222, 224, 229, 231, 233, 238, 240, 242, 247, 249, 251, 254, 257, 26
  • human anti-GPNMB antibodies are Mabl.10.2, Mabl.15.1, Mabl.2.2, Mabl.7.1, Mab2.10.2, Mab2.15.1, Mab2.16.1, Mab2.17.1, Mab2.21.2, Mab2.22.1, Mab2.24.1, Mab2.3.1, Mab2.7.1, and Mab2.8.1. These antibodies have amino acid sequences and nucleic acid sequences encoding them identified in this application as shown in Tables 2A-2D. TABLE 2A Antibod Nucleotide DNA) and Amino Acid (AA Sequences
  • GPNMB-binding human antibodies of the invention comprise germline V heavy chain region VH4-31 or are derived therefrom and have an amino acid sequence of the formula:
  • Xl is E or Q;
  • X2 is T or N
  • X3 is A, F or G
  • X4 is N or G
  • X5 is Y or F
  • X6 is T or S
  • X7 is Q or H
  • X8 is S or N
  • X9 is C, S or Y;
  • XlO is I or T;
  • XI l is K or T;
  • SEQ ID NO:253 is combined with D3-22 or Dl -20. Furthermore the combination of SEQ ID NO:253 with D3-22 or Dl -20 is combined with JH6b or JH4b and in specific embodiments, after affinity maturation these GPNMB-binding human antibodies, for example Mab 1.15.1, Mabl.7.1 and Mab2.22.1, have amino acid sequences SEQ ID NOs:20, 56 and 164 and can be encoded by nucleotide sequences SEQ ID NOs:20, 56 and 164 and can be encoded by nucleotide sequences SEQ
  • H chain CDRl sequences are the germline VH4-31 CDR or affinity matured sequences thereof, of the formula: CDRh GGSIS SX 3 X 4 YX 5 WX 6
  • X3 is A, F or G
  • X4 is N or G
  • an anti-GPNMB antibody of the invention comprise a
  • CDRl sequence selected from the following: SEQ ID NO:22, 58, 166.
  • H chain CDR2 sequences are the germline VH4-31 CDR or affinity matured sequences thereof of the formula:
  • CDR2 Y[YYSGX 8 TYX 9 NPSLIVS Where: X8 is S or N;
  • X9 is C, S or Y; (SEQ ID NO:255).
  • an anti-GPNMB antibody of the invention comprise a CDR2 sequence selected from the following: SEQ ID NO: 24, 60, and 168.
  • the H chain CDR3 sequence is a D3-22, JH6b combination having SEQ ID NO: 170.
  • the H chain CDR3 sequence is a D3-22, JH6b combination having SEQ ID NO: 170.
  • CDR3 sequence is a Dl-20, JH4b combination having SEQ ID NO:26 or 62.
  • GPNMB-binding human antibodies of the invention comprise ge ⁇ nline V heavy chain region VHl -2 or are derived therefrom and include an amino acid sequence of the formula:
  • X2 is A or T;
  • X3 is N or Y;
  • X4 is A or V;
  • X5 is D or G;
  • X6 is A or V;
  • SEQ ID NO:256 is combined with D3-10 or D6-19. Furthermore the combination ov SEQ ID NO:256 with D3-10 or D6-19 is combined with JH4b or JH5b and in specific embodiments, after affinity maturation these GPNMB-binding human antibodies, for example Mab2.3.1 and Mab 2.17.1 have amino acid sequences: SEQ ID NO:128 and 200 and can be encoded by nucleotide sequences SEQ ID NO:127 and 199. Furthermore, in particular embodiments H chain CDRl sequences are the germline VH 1-2 CDR or affinity matured sequences thereof, of the formula:
  • X l is Y or F, (SEQ ID NO:257 )
  • an anti-GPNMB antibody of the invention comprise a CDRl sequence selected from SEQ ID NO: 130 and 202.
  • H chain CDR2 sequences are the germline VH 1-2 CDR or affinity matured sequences thereof of the formula:
  • CDR2 WINPNSGGTX 3 YX 4 QKFQX 5
  • X3 is N or Y
  • X4 is A or V
  • X5 is D or G (SEQ ID NO:258).
  • an anti-GPNMB antibody of the invention comprise a CDR2 sequence selected from SEQ ID NO: 132 and 204.
  • H chain CDR3 sequences are germline D3-10, JH4b combinations or affinity matured sequences thereof, having the amino acid sequence of the formula:
  • Xl is Y or D
  • X2 is Y or F
  • X3 is Y or F
  • an anti-GPNMB antibody of the invention comprise a
  • CDR3 sequence selected from SEQ ID NO: 134 and 206.
  • GPNMB-binding human antibodies of the invention comprise germline V heavy chain region VH2-5 or are derived therefrom and include an amino acid sequence of the formula:
  • X4 is M or V
  • X5 is D or E
  • X6 is A or T
  • X9 is T or R; X lO is M or I; (SEQ ID NO:260 ).
  • SEQ ID NO:260 is combined with D3-9 or D3-16 and furthermore is combined with JH4b.
  • these GPNMB-binding human antibodies for example, Mab 2.8.1 and Mab 1.2.2 have amino acid sequences SEQ ID NO: 38 and 236 and can be encoded by nucleotide sequences SEQ ID NO: 37 and 235.
  • H chain CDRl sequences are the germline VH2-5 CDR or affinity matured sequences thereof, of the formula:
  • X3 is S or G
  • X4 is M or V; (SEQ 1D NO:261 ).
  • an anti-GPNMB antibody of the invention comprise a CDRl sequence selected from SEQ ID NO: 40 and 238.
  • H chain CDR2 sequences are the germline VH2-5 CDR2 or affinity matured sequences thereof of the formula:
  • CDR2 LIYWNDDKX 7 YSPSLX 8 S
  • X7 is R or H
  • X8 is K or R
  • CDR2 sequence selected from SEQ ID NO:42 and 240.
  • H chain CDR3 sequences are germline D3-9, JH4b combinations or affinity matured sequences thereof and include an amino acid sequence of the formula:
  • X2 is Y or F; and X3 is Y or N (SEQ ID NO-.263 ).
  • an anti-GPNMB antibody of the invention comprises a CDR3 amino acid sequence SEQ ID NO:242.
  • H chain CDR3 sequences are germline D3-16, JH4b combinations or affinity matured sequences thereof and include an amino acid sequence of the formula:
  • an anti-GPNMB antibody of the invention comprises a CDR3 amino acid sequence SEQ ID NO: 44.
  • VH3-33 derived anti-GPNMB Antibodies VH3-33 derived anti-GPNMB Antibodies:
  • GPNMB-binding human antibodies of the invention comprise germline V heavy chain region VH3-33 or are derived therefrom and have an amino acid sequence of the formula:
  • X4 is S or N
  • X5 is M or I
  • X6 is Y or F
  • X7 is S or R; XS is T or A;
  • X9 is R or K
  • SEQ ID NO:265 is combined with D3-10 or D4-17 and furthermore with JH4b.
  • these GPNMB- binding human antibodies for example Mab 2.7.1 and Mab2.15.1 have amino acid sequences: SEQ ID NO: 92 and 218 and can be encoded by nucleotide sequences SEQ ID NO:91 and 217.
  • H chain CDRl sequences are the ge ⁇ nline VH3-33 CDR or affinity matured sequences thereof, of the formula:
  • X4 is S or N
  • X5 is M or I; (SEQ ID NO-.266).
  • an anti-GPNMB antibody of the invention comprise a CDRl amino acid sequence selected from SEQ ID NO:94 and 220.
  • H chain CDR2 sequences are the germline VH3-33 CDR2 or affinity matured sequences thereof of the formula:
  • an anti-GPNMB antibody of the invention comprise a CDR2 sequence selected from SEQ ID NO:96 and 222.
  • H chain CDR3 sequences are D3-10, JH4b combinations or affinity matured sequences thereof and include an amino acid sequence of the formula: CDR3: YYYGSGX,
  • a specific embodiment is anti-GPNMB antibody 2.7.1 having a CDR3 amino acid sequence SEQ ID NO:224.
  • H chain CDR3 sequences are D4-17, JH4b combinations or affinity matured sequences thereof and include an amino acid sequence of the formula:
  • a specific embodiment is anti-GPNMB antibody 2.15.1 having a CDR3 amino acid sequence SEQ ID NO: 98.
  • VH3-11 derived anti-GPNMB Antibodies VH3-11 derived anti-GPNMB Antibodies:
  • GPNMB-binding human antibodies of the invention comprise germline V heavy chain region VH3-11 or are derived therefrom and have an amino acid sequence of the formula:
  • Xl is D or S
  • X2 is S or Y
  • X3 is S or T
  • X4 is S or I
  • X5 is T or I
  • SEQ ID NO:270 is combined with D6-13 and furthermore with JH3b.
  • these GPNMB-binding human antibodies for example Mab 2.16.1 have amino acid sequence SEQ ID NO: 110 and can be encoded by nucleotide sequence SEQ ID NO: 109.
  • H chain CDRl sequences are the germline
  • an anti-GPNMB antibody of the invention comprise a
  • H chain CDR2 sequences are the germline VH3-11 CDR2 or affinity matured sequences thereof of the formula:
  • CDR2 YISX 4 SGSX 3 X 6 X 7 YADSVKG
  • X4 is S or I
  • X5 is T or I
  • X6 is T or I
  • X7 is Y or H
  • an anti-GPNMB antibody of the invention comprises a
  • H chain CDR3 sequences are D6-13, JH3b combinations or affinity matured sequences thereof and include an amino acid sequence of the formula:
  • X2 is I or G; (SEQ ID NO:273).
  • a specific embodiment is anti-GPNMB antibody 2.16.1 having a CDR3 amino acid sequence SEQ ID NO: 116.
  • VH3-21 derived anti-GPNMB Antibodies VH3-21 derived anti-GPNMB Antibodies:
  • GPNMB-binding human antibodies of the invention comprise germline V heavy chain region VH3-21 or are derived therefrom and have an amino acid sequence of the formula:
  • Xl is E or Q;
  • X4 is F or L
  • X5 is S or F
  • SEQ ID NO:274 is combined with D 1-26 and furthermore with JH4b.
  • these GPNMB-binding human antibodies for example Mab 2.21.1 have amino acid sequence SEQ ID NO: 146 and can be encoded by nucleotide sequence SEQ ID NO: 145.
  • H chain CDRl sequences are the germline VH3-21 CDRl, SEQ ID NO: 148 or affinity matured sequences thereof.
  • H chain CDR2 sequences are the germline VH3-21 CDR2 or affinity matured sequences thereof of the formula:
  • CDR2 X 5 ISS SSSYIYYADSVKG Where: X5 is S or F;
  • an anti-GPNMB antibody of the invention comprises a
  • H chain CDR3 sequences are D 1-26, JH4b combinations or affinity matured sequences thereof and include an amino acid sequence of the formula:
  • X2 is I or W
  • a specific embodiment is anti-GPNMB antibody 2.21.1 having a CDR3 amino acid sequence SEQ ID NO: 152.
  • GPNMB-binding human antibodies of the invention comprise germline V heavy chain region VH3-30 or are derived therefrom and include an amino acid sequence of the formula:
  • SEQ ID NO:277 is combined with D3-10 and furthermore with JH6b.
  • these GPNMB-binding human antibodies for example Mab 2.10.2 have amino acid sequence SEQ ID NO:74 and can be encoded by nucleotide sequence SEQ ID NO:73.
  • H chain CDRl sequences are the germline VH3-30 CDRl, or affinity matured sequences thereof having an amino acid sequence of the formula: GFXiFS SYGMH
  • Xl is T or A
  • an anti-GPNMB antibody of the invention comprise a
  • H chain CDR2 sequences are the germline V ⁇ 3-30
  • CDR2 or affinity matured sequences thereof of the formula: CDR2: VISYDGX 2 NKyYADSVKG Where: X2 is S or N;
  • an anti-GPNMB antibody of the invention comprises a CDR2 amino acid sequence SEQ ID NO:78.
  • H chain CDR3 sequences are D3-10, JH 6b combinations or affinity matured sequences thereof and include an amino acid sequence of the formula: Where: Xl is I or D; X2 is T or L;
  • a specific embodiment is anti-GPNMB antibody 2.10.2 having a CDR3 amino acid sequence SEQ ID NO:80.
  • VH4-59 derived anti-GPNMB Antibodies VH4-59 derived anti-GPNMB Antibodies:
  • GPNMB-binding human antibodies of the invention comprise germline V heavy chain region VH4-59 or are derived therefrom and include an amino acid sequence of the formula:
  • Xl is G or D
  • X2 is S or N
  • X3 is I or F
  • SEQ ID NO:281 is combined with D6-19 and furthermore with JH4b.
  • these GPNMB-binding human antibodies for example Mab 1.10.2 have amino acid sequence SEQ ID NO:2 and can be encoded by nucleotide sequence SEQ ID NO:1.
  • H chain CDRl sequences are the germline
  • VH4-59 CDRl or affinity matured sequences thereof having an amino acid sequence of the formula:
  • an anti-GPNMB antibody of the invention comprise a CDRl sequence SEQ ID NO:4.
  • H chain CDR2 sequences are the germline VH4-59 CDR2 or affinity matured sequences thereof of the formula:
  • CDR2 YXjYYSGSTNYNPSLKS Where: X3 is I or F; (SEQ ID NO:283).
  • H chain CDR3 sequences are D6-19, JH4b combinations or affinity matured sequences thereof and include an amino acid sequence of the formula: CDR3: X 1 X 2 GW-DY
  • Xl is S or D
  • X2 is S or R
  • SEQ ID NO:284 A specific embodiment is anti-GPNMB antibody 1.10.2 having a CDR3 amino acid sequence SEQ ID NO:8.
  • VH5-51 derived anti-GPNMB Antibodies VH5-51 derived anti-GPNMB Antibodies:
  • GPNMB-binding human antibodies of the invention comprise germline V heavy chain region VH5-51 or are derived therefrom and include an amino acid sequence of the formula:
  • SEQ ID NO:285 is combined with D5-24 and furthermore with JH4b.
  • these GPNMB-binding human antibodies for example Mab 2.24.1 have amino acid sequence SEQ ID NO: 182 and can be encoded by nucleotide sequence SEQ ID NO: 181.
  • H chain CDRl sequences are the germline VH5-51 CDRl, or affinity matured sequences thereof having an amino acid sequence of the formula:
  • X3 is S or N; (SEQ ID NO:286).
  • an anti-GPNMB antibody of the invention comprise a CDRl sequence SEQ ID NO: 184.
  • H chain CDR2 sequences are the germline VH5-51 CDR2 or affinity matured sequences thereof of the formula: CDR2: X 4 IYPX 5 DSDTRYSPSFQG
  • an anti-GPNMB antibody of the invention comprises a CDR2 amino acid sequence SEQ ID NO: 186.
  • H chain CDR3 sequences are D5-24, JH4b combinations or affinity matured sequences thereof and include an amino acid sequence of the formula:
  • CDR3 X,WLQX2--FDY
  • Xl is R or K
  • X2 is L or H
  • a specific embodiment is anti-GPNMB antibody 2.24.1 having a CDR3 amino acid sequence SEQ ID NO: 188.
  • the antibodies of the invention bind an epitope of GPNMB (SEQ ID NO:289), preferably within the mature sequence of GPNMB and more preferably within the extracellular domain (ECD) of GPNMB.
  • Antibodies of the invention bind GPNMB with an affinity of 10 '6 to 10 ⁇ n . Preferably with an affinity of 10 ⁇ 7 or greater and even more preferably 10 " or greater.
  • antibodies described herein bind to GPNMB with very high affinities (Kd), for example a human antibody that is capable of binding GPNMB with a Kd less than, but not limited to, 1(T 7 , 10 "8 , 1(T 9 , 10 "10 , 1(T 11 , 1(T 12 , ICT 13 or 10 "14 M, or any range or value therein.
  • Kd very high affinities
  • Affinity and/or avidity measurements can be measured by KinExA ® and/or BIACORE ® , as described herein.
  • antibodies of the invention bind to GPNMB with Kds ranging from 50 to 150 pM.
  • Epitope mapping and secondary and tertiary structure analyses can be carried out to identify specific 3D structures assumed by the disclosed antibodies and their complexes with antigens (see, e.g., Epitope Mapping Protocols, ed. Morris, Humana Press, 1996). Such methods include, but are not limited to, X-ray crystallography (Biochem. Exp. Biol, 11: 7-13, 1974) and computer modeling of virtual representations of the presently disclosed antibodies (Fletterick et al. (1986) Computer Graphics and Molecular Modeling, in Current Communications in Molecular Biology, Cold Spring Harbor Laboratory, Cold Spring Harbor, N. Y).
  • the specific part of the protein immunogen recognized by antibody may be determined by assaying the antibody reactivity to parts of the protein, for example an N terminal and C terminal half. The resulting reactive fragment can then be further dissected, assaying consecutively smaller parts of the immunogen with the antibody until the minimal reactive peptide is defined.
  • the binding specificity, that is the epitope, of anti-GPNMB antibodies of the invention may be determined by subjecting GPNMB immunogen to SDS-PAGE either in the absence or presence of a reduction agent and analyzed by immunoblotting. Epitope mapping may also be performed using SELDI. SELDI ProteinChip® (LumiCyte) arrays used to define sites of protein-protein interaction. GPNMB protein antigen or fragments thereof may be specifically captured by antibodies covalently immobilized onto the PROTEINCHIP array surface. The bound antigens may be detected by a laser-induced desorption process and analyzed directly to determine their mass.
  • the epitope recognized by anti-GPNMB antibodies described herein may be determined by exposing the PROTEINCHIP Array to a combinatorial library of random peptide 12-mer displayed on Filamentous phage (New England Biolabs). Antibody-bound phage are eluted and then amplified and taken through additional binding and amplification cycles to enrich the pool in favor of binding sequences. After three or four rounds, individual binding clones are further tested for binding by phage ELISA assays performed on antibody-coated wells and characterized by specific DNA sequencing of positive clones. Derivatives
  • CDRs in such antibodies are not limited to the specific sequences of H and L variable domains identified in Table 1 and may include variants of these sequences that retain the ability to specifically bind GPNMB. Such variants may be derived from the sequences listed in Table 1 by a skilled artisan using techniques well known in the art. For example, amino acid substitutions, deletions, or additions, can be made in the FRs and/or in the CDRs. While changes in the FRs are usually designed to improve stability and immunogenicity of the antibody, changes in the CDRs are typically designed to increase affinity of the antibody for its target. Variants of FRs also include naturally occurring immunoglobulin allotypes.
  • affinity-increasing changes may be determined empirically by routine techniques that involve altering the CDR and testing the affinity of the antibody for its target. For example, conservative amino acid substitutions can be made within any one of the disclosed CDRs.
  • Various alterations can be made according to the methods described in the art (Antibody Engineering, 2.sup.nd ed., Oxford University Press, ed. Borrebaeck, 1995). These include but are not limited to nucleotide sequences that are altered by the substitution of different codons that encode a functionally equivalent amino acid residue within the sequence, thus producing a "silent" change.
  • the nonpolar amino acids include alanine, leucine, isoleucine, valine, proline, phenylalanine, tryptophan, and methionine.
  • the polar neutral amino acids include glycine, serine, threonine, cysteine, tyrosine, asparagine, and glutamine.
  • the positively charged (basic) amino acids include arginine, lysine, and histidine.
  • the negatively charged (acidic) amino acids include aspartic acid and glutamic acid. Substitutes for an amino acid within the sequence may be selected from other members of the class to which the amino acid belongs (see Table 3).
  • any native residue in the polypeptide may also be substituted with alanine (Acta Physiol. Scand. Suppl. 643:55-67, 1998; Adv. Biophys. 35:1-24, 1998).
  • Derivatives and analogs of antibodies of the invention can be produced by various techniques well known in the art, including recombinant and synthetic methods (Maniatis (1990) Molecular Cloning, A Laboratory Manual, 2.sup.nd ed., Cold Spring Harbor Laboratory, Cold Spring Harbor, N. Y., and Bodansky et al. (1995) The Practice of Peptide Synthesis, 2.sup.i ⁇ d ed., Spring Verlag, Berlin, Germany).
  • Preferred amino acid substitutions are those which: (1) reduce susceptibility to proteolysis, (2) reduce susceptibility to oxidation, (3) alter binding affinity for forming protein complexes, (4) alter binding affinities, and (4) confer or modify other physicochemical or functional properties of such analogs.
  • Analogs can include various muteins of a sequence other than the naturally-occurring peptide sequence. For example, single or multiple amino acid substitutions (preferably conservative amino acid substitutions) may be made in the naturally-occurring sequence (preferably in the portion of the polypeptide outside the domain(s) forming intermolecular contacts).
  • a conservative amino acid substitution should not substantially change the structural characteristics of the parent sequence ⁇ e.g., a replacement amino acid should not tend to break a helix that occurs in the parent sequence, or disrupt other types of secondary structure that characterizes the parent sequence).
  • a replacement amino acid should not tend to break a helix that occurs in the parent sequence, or disrupt other types of secondary structure that characterizes the parent sequence.
  • Examples of art-recognized polypeptide secondary and tertiary structures are described in the art (for example, Proteins, Structures and Molecular Principles (Creighton, Ed., W. H. Freeman and Company, New York (1984)).
  • a method for making an H variable domain which is an amino acid sequence variant of an H variable domain of the invention comprises a step of adding, deleting, substituting, or inserting one or more amino acids in the amino acid sequence of the presently disclosed H variable domain, optionally combining the H variable domain thus provided with one or more L variable domains, and testing the H variable domain or H variable/L variable combination or combinations for specific binding to GPNMB or and, optionally, testing the ability of such antigen-binding domain to modulate GPNMB activity.
  • the L variable domain may have an amino acid sequence that is identical or is substantially as set out according to Table 1.
  • An analogous method can be employed in which one or more sequence variants of a L variable domain disclosed herein are combined with one or more H variable domains.
  • a further aspect of the disclosure provides a method of preparing antigen-binding fragment that specifically binds with GPNMB.
  • the method comprises: (a) providing a starting repertoire of nucleic acids encoding a H variable domain that either includes a
  • CDR3 to be replaced or lacks a CDR3 encoding region; (b) combining the repertoire with a donor nucleic acid encoding an amino acid sequence substantially as set out herein for a H variable CDR3 such that the donor nucleic acid is inserted into the CDR3 region in the repertoire, so as to provide a product repertoire of nucleic acids encoding a H variable domain; (c) expressing the nucleic acids of the product repertoire; (d) selecting a binding fragment specific for GPNMB; and (e) recovering the specific binding fragment or nucleic acid encoding it.
  • a L variable CDR3 of the invention is combined with a repertoire of nucleic acids encoding a L variable domain, which either include a CDR3 to be replaced or lack a CDR3 encoding region.
  • the donor nucleic acid may be selected from nucleic acids encoding an amino acid sequence substantially as set out in SEQ ID NOs: 2, 20, 38, 56, 74, 92, 110, 128, 146, 164, 182, 200, 218, 236, 253, 256, 260, 265, 270, 274, 277, 281, 285, 1 1 , 29, 47, 65, 83, 101, 1 19, 137,
  • a sequence encoding a CDR of the invention ⁇ e.g., CDR3) may be introduced into a repertoire of variable domains lacking the respective CDR ⁇ e.g., CDR3), using recombinant DNA technology, for example, using methodology described by Marks et al. ⁇ Bio/Technology (1992) 10: 779-783).
  • consensus primers directed at or adjacent to the 5' end of the variable domain area can be used in conjunction with consensus primers to the third framework region of human H variable genes to provide a repertoire of H variable domains lacking a CDR3.
  • the repertoire may be combined with a CDR3 of a particular antibody.
  • the CDR3-derived sequences may be shuffled with repertoires of H variable or L variable domains lacking a CDR3, and the shuffled complete H variable or L variable domains combined with a cognate L variable or H variable domain to make the GPNMB specific antibodies of the invention.
  • the repertoire may then be displayed in a suitable host system such as the phage display system such as described in WO92/01047 so that suitable antigen-binding fragments can be selected.
  • Analogous shuffling or combinatorial techniques may be used (e.g. Stemmer, Nature
  • Another method that may be used is to direct mutagenesis to CDRs of H variable or L variable genes ⁇ Proc. Nat. Acad. ScL U.S.A. (1994) 91: 3809-3813; J. MoI. Biol. (1996) 263: 551-567).
  • one or more, or all three CDRs may be grafted into a repertoire of H variable or L variable domains, which are then screened for an antigen-binding fragment specific for GPNMB.
  • a portion of an immunoglobulin variable domain will comprise at least one of the CDRs substantially as set out herein and, optionally, intervening framework regions as set out herein.
  • the portion may include at least about 50% of either or both of FRl and FR4, the 50% being the C-terminal 50% of FRl and the N-terminal 50% of FR4. Additional residues at the N-terminal or C-terminal end of the substantial part of the variable domain may be those not normally associated with naturally occurring variable domain regions.
  • construction of antibodies by recombinant DNA techniques may result in the introduction of N- or C-terminal residues encoded by linkers introduced to facilitate cloning or other manipulation steps.
  • variable domains include immunoglobulin heavy chain constant regions, other variable domains (for example, in the production of diabodies), or proteinaceous labels as discussed in further detail below.
  • linkers to join variable domains include immunoglobulin heavy chain constant regions, other variable domains (for example, in the production of diabodies), or proteinaceous labels as discussed in further detail below.
  • embodiments illustrated in the Examples comprise a "matching" pair of H variable and L variable domains
  • alternative embodiments may comprise antigen-binding fragments containing only a single CDR from either L variable or H variable domain. Either one of the single chain specific binding domains can be used to screen for complementary domains capable of forming a two- domain specific antigen-binding fragment capable of, for example, binding to GPNMB.
  • the screening may be accomplished by phage display screening methods using the so-called hierarchical dual combinatorial approach disclosed in WO92/01047, in which an individual colony containing either an H or L chain clone is used to infect a complete library of clones encoding the other chain (L or H) and the resulting two-chain specific binding domain is selected in accordance with phage display techniques as described.
  • Anti-GPNMB antibodies described herein can be linked to another functional molecule, e.g., another peptide or protein (albumin, another antibody, etc.), toxin, radioisotope, cytotoxic or cytostatic agents.
  • the antibodies can be linked by chemical cross-linking or by recombinant methods.
  • the antibodies may also be linked to one of a variety of nonproteinaceous polymers, e.g. , polyethylene glycol, polypropylene glycol, or polyoxyalkylenes, in the manner set forth in U.S. Pat. Nos. 4,640,835; 4,496,689; 4,301,144; 4,670,417; 4,791,192; or 4,179,337.
  • the antibodies can be chemically modified by covalent conjugation to a polymer, for example, to increase their circulating half-life.
  • exemplary polymers and methods to attach them are also shown in U.S. Pat. Nos. 4,766,106; 4,179,337; 4,495,285, and 4,609,546.
  • the disclosed antibodies may also be altered to have a glycosylation pattern that differs from the native pattern.
  • one or more carbohydrate moieties can be deleted and/or one or more glycosylation sites added to the original antibody.
  • Addition of glycosylation sites to the presently disclosed antibodies may be accomplished by altering the amino acid sequence to contain glycosylation site consensus sequences known in the art.
  • Another means of increasing the number of carbohydrate moieties on the antibodies is by chemical or enzymatic coupling of glycosides to the amino acid residues of the antibody (WO 87/05330; CRC Crit. Rev. Biochem., 22: 259-306, 1981). Removal of any carbohydrate moieties from the antibodies may be accomplished chemically or enzymatically (Arch. Biochem.
  • the antibodies may also be tagged with a detectable, or functional, label.
  • Detectable labels include radiolabels such as 131 I or 99 Tc, which may also be attached to antibodies using conventional chemistry.
  • Detectable labels also include enzyme labels such as horseradish peroxidase or alkaline phosphatase.
  • Detectable labels further include chemical moieties such as biotin, which may be detected via binding to a specific cognate detectable moiety, e.g., labeled avidin.
  • the valency of the antibodies may be custom designed to affect affinity and avidity, retention time at binding sites (see e.g. Am H. Pathol, 2002 160:1597-1608; J. Med. Chem. 2002 45:2250-2259; Br. J. Cancer 2002 86:1401-1410; Biomol. Eng. 2001 18:95-108; Int J. Cancer 2002 100:367-374).
  • bispecific binding reagents may be designed based upon the GPNMB specific sequences of the invention (Biomol. Eng.2001 18:31-40).
  • a bispecific or bifunctional antibody is an artificial hybrid antibody having two different heavy/light chain pairs and two different binding sites.
  • Bispecific antibodies can be produced by a variety of methods including fusion of hybridomas or linking of Fab' fragments (Clin. Exp. Immunol. 1990, 79: 315-321;, J. Immunol. 199,2148:1547-1553).
  • bispecific antibodies can be generated comprising a specificity to GPNMB and a second specificity to a second molecule using techniques that are well known (Immunol Methods 1994,4:72-81; Wright and Harris, supra. ; Traunecker et all 992 Int. J. Cancer (Suppl.) 7:51-52). Bispecific antibodies prepared in this manner selectively kill cells expressing GPNMB.
  • Antibodies in which CDR sequences differ only insubstantially from those set out in SEQ ID NOs: 4, 6, 8, 13, 15, 17, 22, 24, 26, 31, 33, 35, 40, 42, 44, 49, 51, 53, 58, 60, 62, 67, 69, 71, 76, 78, 80, 85, 87, 89, 94, 96, 98, 103, 105, 107, 1 12, 1 14, 116, 121, 123, 125, 130, 132, 134, 139, 141, 143, 148, 150, 152, 157, 159, 161, 166, 168, 170, 175, 177, 179, 184, 186, 188, 193, 195, 197, 202, 204, 206, 21 1, 213, 215, 220, 222, 224, 229, 231, 233, 238, 240, 242, 247, 249 and 251.
  • Changes to FRs include, but are not limited to engineering certain framework residues that are important for antigen contact or for stabilizing the binding site, e.g., changing the class or subclass of the constant region, changing specific amino acid residues which might alter the effector function such as Fc receptor binding (U.S. Pat. Nos. 5,624,821; 5,648,260;Lund et al. (1991) J. Immiin. 147: 2657 -2662 ;Morgan et al. (1995) Immunology 86: 319-324), or changing the species from which the constant region is derived.
  • Fc receptor binding U.S. Pat. Nos. 5,624,821; 5,648,260;Lund et al. (1991) J. Immiin. 147: 2657 -2662 ;Morgan et al. (1995) Immunology 86: 319-324
  • the present disclosure further provides isolated nucleic acids encoding the disclosed antibodies.
  • the nucleic acids may comprise DNA or RNA and may be wholly or partially synthetic or recombinant.
  • Reference to a nucleotide sequence as set out herein encompasses a DNA molecule with the specified sequence, and encompasses a RNA molecule with the specified sequence in which U is substituted for T, unless context requires otherwise.
  • the nucleic acids provided herein comprise a coding sequence for a CDR, a H variable domain, and/or a L variable domain disclosed herein.
  • the present disclosure also provides constructs in the fo ⁇ n of plasmids, vectors, phagemids, transcription or expression cassettes which comprise at least one nucleic acid encoding a CDR, a H variable domain, and/or a L variable domain disclosed here.
  • the disclosure further provides a host cell which comprises one or more constructs as above.
  • nucleic acids encoding any CDR (CDRl, CDR2, CDR3 from either the H or L variable domain), H variable or L variable domain, as well as methods of making of the encoded products.
  • the method comprises expressing the encoded product from the encoding nucleic acid. Expression may be achieved by culturing under appropriate conditions recombinant host cells containing the nucleic acid. Following production by expression, a H variable or L variable domain, or specific binding member may be isolated and/or purified using any suitable technique, then used as appropriate.
  • Antigen-binding fragments, FI variable and/or L variable domains and encoding nucleic acid molecules and vectors may be isolated and/or purified from their natural environment, in substantially pure or homogeneous form, or, in the case of nucleic acid, free or substantially free of nucleic acid or genes of origin other than the sequence encoding a polypeptide with the required function.
  • Systems for cloning and expression of a polypeptide in a variety of different host cells are well known in the art including cells suitable for producing antibodies ⁇ Gene Expression Systems, Academic Press, eds. Fernandez et al., 1999). Briefly, suitable host cells include bacteria, plant cells, mammalian cells, and yeast and baculovirus systems.
  • Mammalian cell lines available in the art for expression of a heterologous polypeptide include Chinese hamster ovary (CHO) cells, HeLa cells, baby hamster kidney cells, NSO mouse myeloma cells, and many others.
  • a common bacterial host is E, coli.
  • Any protein expression system compatible with the invention may be used to produce the disclosed antibodies.
  • Suitable expression systems also include transgenic animals ⁇ Gene Expression Systems, Academic Press, eds. Fernandez et at, 1999).
  • Suitable vectors can be chosen or constructed, so that they contain appropriate regulatory sequences, including promoter sequences, terminator sequences, polyadenylation sequences, enhancer sequences, marker genes and other sequences as appropriate.
  • Vectors may be plasmids or viral, e.g., phage, or phagemid, as appropriate ⁇ see Sambrook et al., Molecular Cloning: A Laboratory Manual, Lsup.nd ed., Cold Spring Harbor Laboratory? Press, 1989). Many known techniques and protocols for manipulation of nucleic acid, for example, in preparation of nucleic acid constructs, mutagenesis, sequencing, introduction of DNA into cells and gene expression, and analysis of proteins, are known in the art ⁇ Current Protocols in Molecular Biology, 2. sup. nd Edition, eels. Ausubel et al, John Wiley & Sons, 1992).
  • the invention also provides a host cell comprising a nucleic acid as disclosed herein.
  • a still further aspect provides a method comprising introducing such nucleic acid into a host cell.
  • the introduction may employ any available technique.
  • suitable techniques may include calcium phosphate transfection, DEAE-Dextran, electroporation, liposome-mediated transfection and transduction using retrovirus or other virus, e.g. , vaccinia or, for insect cells, baculovirus.
  • suitable techniques may include calcium chloride transformation, electroporation and transfection using bacteriophage.
  • the introduction of the nucleic acid into the cells may be followed by causing or allowing expression from the nucleic acid, e.g., by culturing host cells under conditions for expression of the gene. Immunoconjugates
  • the antibodies of the invention can be used as a targeting agent for delivery of another therapeutic or a cytotoxic agent to a cell expressing GPNMB.
  • the method includes administering an anti-GPNMB antibody coupled to a therapeutic or a cytotoxic agent or under conditions that allow binding of the antibody to GPNMB.
  • Anti-GPNMB antibodies are conjugated to a therapeutic agent, such as a cytotoxic compound, such that the resulting immimoconjugate exerts a cytotoxic or cytostatic effect on a GPNMB expressing cell.
  • a therapeutic agent such as a cytotoxic compound
  • Particularly suitable moieties for conjugation to antibodies are chemotherapeutic agents, prodrug converting enzymes or toxins.
  • an anti- GPNMB antibody can be conjugated to a cytotoxic agent such as a chemotherapeutic agent (see infra) or a toxin (e.g. abrin, ricin A, pseudomonas exotoxin, or diphtheria toxin).
  • anti-GPNMB antibody may be conjugated to a pro-drug converting enzyme.
  • the pro-drug converting enzyme can be recombinantly fused to the antibody or derivative thereof or chemically conjugated thereto using known methods.
  • Examplary pro-drug converting enzymes are carboxypeptidase G2, ⁇ -glucuronidase, penicillin- V-amidase, penicillin-G-amidase, ⁇ -lactamase, ⁇ -glucosidase, nitroreductase and carboxypeptidase A.
  • Any agent that exerts a therapeutic effect on GPNMB expressing cells can be used as an agent for conjugation to an anti-GPNMB antibody of the invention.
  • cytotoxic agents include, for example, antitubulin agents, auristatins, DNA minor groove binders, NDA replication inhibitiors, alkylating agents (e.g., platinum complexes such as cis-plantin, mono(platinum), bis(platinum) and tri-nuclear platinum complexes and carboplatin), anthracyclines, antiboiotics, antifolates, antimetabilites, chemotherapy sensitizers, duocarmycins, etoposides, fluorinated purimidines, ionophores, lexitropsins, nitrosoureas, platinols, pre-forming compounds, purine antimetabolites, puromcins, radiation sensitizers, steroids, taxanes, topoisomerase inhibitors, vinca alkaloids, or the like.
  • alkylating agents e.g., platinum complexes such as cis-plantin, mono(platinum), bis(platinum
  • the therapeutic agent can be a cytotoxic agent.
  • Suitable cytotoxic agents include, for example, dolastatins (e.g. auristatin E, AFP, MMAF, MMAE), DNA minor groove binders (e.g., enediynes and lexitropsins), cuocarmycins, taxanes (e.g., paclitaxel and docetaxel), puromycins, vinca alkaloids, CC- 1065, SN-38, topotecan, morpholino- doxorubicin, rhizoxin, cyanomorpholino-doxorubicin, echinomycin, combretastatin, netropsin, epothilone A and B, estramustine, cryptophysins, cemadotin, maytansinoids, discode ⁇ nolide, eleutherobin, and mitoxantrone.
  • dolastatins e.g. auristat
  • the cytotoxic or cytostatic agent is auristatin E (dolastatin- 10) or a derivative thereof (e.g. an ester formed between auristatin E and a keto acid).
  • auristatin E diolastatin- 10
  • Other typical auristatin derivatives include AFP, MMAR, and MMAE. The synthesis and structure of auristatin E and its derivates are described in U.S. Patent Application
  • anti-GPNMB antibody 1.15.1 was coupled to monomethylauristatin E via intracellular protease-sensitive valine-citruUine peptide linker (vcMMAE).
  • vcMMAE intracellular protease-sensitive valine-citruUine peptide linker
  • anti-GPNMB antibodies binding to GPNMB expressing cells are internalized and accumulate in the cell.
  • anti-GPNMB antibody immunoconjugates accumulate in GPNMB expressing cells.
  • the agent is preferentially active.
  • anti GPNMB immunoconjugates are not internalized and the drug is effective to deplete or inhibit GPNMB expressing cells by binding to the cell membrane.
  • the therapeutic agent can be conjugated in a manner that reduces its activity unless it is cleaved off the antibody (e.g.
  • the agent can be attached to the antibody or derivative thereof with a cleavable linker that is sensitive to cleavage in the intracellular environment of the target but is not substantially sensitive to the extracellular environment, such that the conjugate is cleaved from the antibody or derivative thereof when it is internalized by the GPNMB expressing cell (e.g. in the endosomal or, for example by virtue of pH sensitivity or protease sensitivity, in the lysosomal environament or in a caveolea).
  • a therapeutic agent of the immunoconjugate can be charged relative to the plasma membrane (e.g. polarized or net charge relative to the plasma membrane), thereby further minimizing the ability of the agent to cross the plasma membrane once internalized by a cell.
  • the anti-GPNMB antibody immunoconjugate can comprise a linker region between the therapeutic agent and the antibody.
  • the linker can be cleavable under intracellular conditions, such that cleavage of the linker releases the therapeutic agent from the antibody in the intracellular environment.
  • the linker can be, e.g. a peptidyl linker that is cleaved by an intracellular peptidase or protease enzyme, including but not limited to a lysosomal or endosomal protease. Often the peptidyl linker is at least two amino acids long or at least three amino acids long.
  • Cleaving agnets can include cathepsins and D and plasmin, all of which are known to hydrolyze dipeptide drug derivative s resulting in the release of active drug inside target cells (see Dubowchik and Walker, 1999 Pharm. Therapuetics 83:67-123).
  • Other linkers are described e.g. in U.S. Patent No. 6,214,345.
  • Linkers can be pH-sensitive can often be hydrolizable under acidic conditions such as is found in the lysosome (see e.g. U.S. Patent Nos. 5,122,368; 5,824,805; 5,622,929; Dubowchik and Walker, 1999 Pharm. Therapuetics 83:67-123; Neville et ah, 1989 BIoI. Chem. 264:14653-14661).
  • Such linkers are relatively stable under neutral pH conditions, such as those in the blood, but are unstable at below pH 5.5 or 5.0, the pH of the lysosome.
  • Linkers can be cleavable under reducing conditions (e.g.
  • the linker can be a malonate linker (Johnson et al, 1995, Anticancer Res. 15: 1387-93), a maleimidobenzoly linker (lau et al, 1995, Bioorg- Med-Chem. 3(10):1299-1304) or a 3'-N-amide analog (Lau et al, 1995, Bioorg-Med- Chem.3(10):1305-1312).
  • the antibodies of the invention can act as either agonists or antagonists of GPNMB, depending on the methods of their use.
  • the antibodies can be used to prevent, diagnose, or treat medical disorders in a subject, especially in humans.
  • Antibodies of the invention can also be used for isolating GPNMB or GPNMB-expi'essing cells.
  • the antibodies can be used to treat a subject at risk of or susceptible to a disorder or having a disorder associated with aberrant GPNMB expression or function.
  • Antibodies of the invention can be used to detect GPNMB in such subjects.
  • the present invention provides methods for treating and/or preventing a disease or disorder associated with overexpression of GPNMB and/or cell hyperproliferative disorders, particularly cancer, in a subject comprising administering an effective amount of a composition that can target cells expressing GPNMB, and inhibiting the GPNMB expression or function, and/or having therapeutic or prophylactic effects on the hyperproliferative cell disease.
  • the method of the invention comprises administering to a subject a composition comprising an immunoconjugate that comprises an antibody of the invention and a cytotoxic agent against the hyperproliferative cell disease.
  • the method of the invention comprises administering to a subject in need thereof a composition comprising a naked IgGl antibody of the invention and one or more immunomodulators.
  • the method of the invention comprises administering to a subject in need thereof a composition comprosing a single chain Fv antibody (anti-GPNMB) conjugated to a cytotoxic agent, or a composition comprising a bispecific antibody that have a single chain anti-GPNMB antibody component and a anti-CD3 antibody component.
  • the hyperproliferative cell disease is cancer. More preferably, the cancer is melanoma, or a cancer of the CNS system, such as astrocytoma, glioblastoma, medulloblastoma, or neoplastic meningitis.
  • the present invention provides therapies comprising administering one of more antibodies of the invention and compositions comprising said antibodies to a subject, preferably a human subject, for preventing and/or treating a disorder characterized by or associated with aberrant expression and/or activity of GPNMB or a symptom thereof.
  • the invention provides a method of preventing or treating a disorder characterized by or associated with aberrant expression and/or activity of GPNMB or a symptom thereof, said method comprising administering to a subject in need thereof an effective amount of one or more antibodies of the invention.
  • an effective amount of one or more immunoconjugates comprising one or more antibodies of the invention is administered to a subject in need thereof to prevent or treat a disorder characterized by or associated with aberrant expression and/or activity of GPNMB or a symptom thereof.
  • the invention also provides methods of preventing or treating a disorder characterized by or associated with aberrant expression and/or activity of GPNMB or a symptom thereof, said methods comprising administering to a subject in need thereof one or more of the antibodies of the invention and one or more therapies (e.g., one or more prophylactic or therapeutic agents) other than antibodies of the invention.
  • the prophylactic or therapeutic agents of the combination therapies of the invention can be administered sequentially or concurrently.
  • the combination therapies of the invention comprise an effective amount of one or more antibodies of the invention and an effective amount of at least one other therapy (e.g., prophylactic or therapeutic agent) which has a different mechanism of action than said antibodies.
  • the combination therapies of the present invention improve the prophylactic or therapeutic effect of one or more antibodies of the invention by functioning together with the antibodies to have an additive or synergistic effect.
  • the combination therapies of the present invention reduce the side effects associated with the therapies (e.g., prophylactic or therapeutic agents).
  • the prophylactic or therapeutic agents of the combination therapies can be administered to a subject, preferably a human subject, in the same pharmaceutical composition.
  • the prophylactic or therapeutic agents of the combination therapies can be administered concurrently to a subject in separate pharmaceutical compositions.
  • the prophylactic or therapeutic agents may be administered to a subject by the same or different routes of administration.
  • a pharmaceutical composition comprising one or more antibodies of the invention described herein is administered to a subject, preferably a human, to prevent and/or treat a disorder characterized by or associated with aberrant expression and/or activity of GPNMB or a symptom thereof.
  • pharmaceutical compositions of the invention may also comprise one or more therapies (e.g., prophylactic or therapeutic agents), other than antibodies of the invention.
  • the antibodies of the invention may also be used to detect the presence of GPNMB in biological samples (in diagnostic methods or use as an efficacy marker).
  • the amount of GPNMB detected may be correlated with the expression level of GPNMB, which, in turn, is correlated with the disease, tumor type, tumor burden or stage using methods known in the art (see for example recommendations of the AAPS Ligand Binding Assay Bioanalytical Focus Group (LBABFG) Pharm Res. 2003 Nov;20(ll): 1885-900).
  • Detection methods that employ antibodies are well known in the art and include, for example, ELISA, radioimmunoassay, immunoblot, Western blot, IHC, immunofluorescence, immunoprecipitation.
  • the antibodies may be provided in a diagnostic kit that incorporates one or more of these techniques to detect GPNMB.
  • a kit may contain other components, packaging, instructions, or other material to aid the detection of the protein.
  • the antibodies of the invention are conjugated to a radioactive isotope, and are injected to a subject to detect cells that overexpressing GPNMB.
  • the antibodies are intended for diagnostic purposes, it may be desirable to modify them, for example, with a ligand group (such as biotin) or a detectable marker group (such as a fluorescent group, a radioisotope or an enzyme).
  • a detectable label such as a fluorescent group, a radioisotope or an enzyme.
  • the antibodies of the invention may be labeled using conventional techniques. Suitable detectable labels include, for example, fiuorophores, chromophores, radioactive atoms, electron-dense reagents, enzymes, and ligands having specific binding partners. Enzymes are typically detected by their activity. For example, horseradish peroxidase can be detected by its ability to convert tetramethylbenzidine (TMB) to a blue pigment, quantifiable with a spectrophotometer.
  • TMB tetramethylbenzidine
  • binding partners include, but are not limited to, biotin and avidin or streptavidin, IgG and protein A, and the numerous receptor-ligand couples known in the art. Other permutations and possibilities will be readily apparent to those of ordinary skill in the art, and are considered as equivalents within the scope of the instant invention.
  • Antibodies of the invention can be used in screening methods to identify inhibitors of GPNMB effective as therapeutics.
  • a first binding mixture is formed by combining GPNMB and an antibody of the invention; and the amount of binding in the first binding mixture (M 0 ) is measured.
  • a second binding mixture is also formed by combining GPNMB, the antibody, and the compound or agent to be screened, and the amount of binding in the second binding mixture (Mi) is measured.
  • a compound to be tested may be another anti-GPNMB antibody. The amounts of binding in the first and second binding mixtures are then compared, for example, by calculating the Mi /Mo ratio.
  • the compound or agent is considered to be capable of modulating a GPNMB-associated responses if a decrease in binding in the second binding mixture as compared to the first binding mixture is observed.
  • the formulation and optimization of binding mixtures is within the level of skill in the art, such binding mixtures may also contain buffers and salts necessary to enhance or to optimize binding, and additional control assays may be included in the screening assay of the invention.
  • Compounds found to reduce the GPNMB-antibody binding by at least about 10% ⁇ i.e., Mj/M 0 0 .9), preferably greater than about 30% may thus be identified and then, if desired, secondarily screened for the capacity to ameliorate a disorder in other assays or animal models as described below.
  • the strength of the binding between GPNMB and an antibody can be measured using, for example, an enzyme-linked immunoadsorption assay (ELISA), radioimmunoassay (RIA), surface plasmon resonance- based technology (e.g., Biacore), all of which are techniques well known in the art.
  • ELISA enzyme-linked immunoadsorption assay
  • RIA radioimmunoassay
  • Biacore surface plasmon resonance- based technology
  • the amount of a prophylactic or therapeutic agent or a composition of the invention which will be effective in the prevention and/or treatment of a disorder associated with or characterized by aberrant expression and/or activity of GPNMB can be determined by standard clinical methods.
  • the dosage of the composition which will be effective in the treatment and/or prevention of cancer can be determined by administering the composition to an animal model.
  • in vitro assays may optionally be employed to help identify optimal dosage ranges.
  • Preliminary doses as, for example, determined according to animal tests, and the scaling of dosages for human administration is performed according to art-accepted practices. Toxicity and therapeutic efficacy can be determined by standard pharmaceutical procedures in cell cultures or experimental animals.
  • the data obtained from the cell culture assays or animal studies can be used in formulating a range of dosage for use in humans.
  • Therapeutically effective dosages achieved in one animal model can be converted for use in another animal, including humans, using conversion factors known in the art (see, e.g., Freireich et al. (1966) Cancer Chemother. Reports, 50(4): 219-244).
  • Selection of the preferred effective dose can be determined (e.g., via clinical trials) by a skilled artisan based upon the consideration of several factors which will be known to one of ordinary skill in the art. Such factors include the disease to be treated or prevented, the symptoms involved, the patient's body mass, gender, immune status and other factors known by the skilled artisan to reflect the accuracy of administered pharmaceutical compositions. Suitable regimens can be selected by one skilled in the art by considering such factors and by following, for example, dosages reported in literature and recommended in the Physician's Desk Reference (59th ed., 2005).
  • Effective doses may be extrapolated from dose-response curves derived from in vitro or animal model test systems.
  • the dosage of an antibody or an immunoconjugate comprising an antibody of the invention administered to prevent and/or treat a disorder associated with or characterized by aberrant expression and/or activity of GPNMB ⁇ e.g., cancer) in a patient is 30 mg/kg or less, 25 mg/kg or less, 20 mg/kg or less, 15 nig/kg or less, preferably 12 mg/kg or less, 11 mg/kg or less, 10 mg/kg or less, 9 mg/kg or less, 8 mg/kg or less, 7 mg/kg or less, 6 mg/kg or less, 5 mg/kg or less, 4 mg/kg or less, 3 mg/kg or less, 2 mg/kg or less, or 1 mg/kg or less of a patient's body weight.
  • the dosage of an antibody or an immunoconjugate of the invention administered to prevent and/or treat a disorder associated with or characterized by aberrant expression and/or activity of GPNMB ⁇ e.g., cancer) in a patient is a unit dose of about 0.01 mg/kg to about 20 mg/kg, about 0.1 mg/kg to about 10 mg/kg, about 0.1 mg/kg to about 8 mg/kg, about 0.1 mg/kg to about 7 mg/kg, about 0.1 mg/kg to about 6 mg/kg, about 0.1 mg/kg to about 5 mg/kg, about 0.1 mg/kg to about 4 mg/kg, preferably, about 0.1 mg/kg to about 3 mg/kg, about 0.2 mg/kg to 3 mg/kg, about 0.3 mg/kg to about 3 mg/kg, about 0.4 mg/kg to about 3 mg/kg, about 0.6 mg/kg to about 3 mg/kg, about 0.8 mg/kg to about 3 mg/kg, about 0.1 mg/kg to 2 mg/kg, about 0.1 mg/kg to 1
  • the dosage of an antibody or an immunoconjugate comprising an antibody of the invention administered to prevent and/or treat a disorder associated with or characterized by aberrant expression and/or activity of GPNMB ⁇ e.g., cancer) in a patient is a unit dose of about 0.1 mg/kg, about 0.2 mg/kg, about 0.4 mg/kg, about 0.6 mg/kg, about 0.8 mg/kg, about 1.1 mg/kg, or about 1 mg/kg.
  • a subject is administered one or more doses of an effective amount of one or more antibodies or imunoconjugates of the invention to prevent and/or treat a disorder associated with or characterized by aberrant expression and/or activity of GPNMB, wherein the dose of an effective amount of said antibodies, immunoconjugates, compositions, or combination therapies reduces and/or inhibits proliferation of cancerous cells by at least 20% to 25%, preferably at least 25% to 30%, at least 30% to 35%, at least 35% to 40%, at least 40% to 45%, at least 45% to 50%, at least 50% to 55%, at least 55% to 60%, at least 60% to 65%, at least 65% to 70%, at least 70% to 75%, at least 75% to 80%, at least 80 to 85%, at least 85% to 90%, at least 90% to 95%, or at least 95% to 98% relative to a control such as PBS in an in vitro and/or in vivo assay well-known in the art.
  • a control such as PBS in an in vitro and/
  • a subject is administered one or more doses of an effective amount of one or more antibodies or immunoconjugates of the invention to prevent and/or treat a disorder associated with or characterized by aberrant expression and/or activity of GPNMB, wherein the dose of an effective amount achieves a serum titer of at least 0.1 ⁇ g/mL, at least 0.5 ⁇ g/mL, at least 1 ⁇ g/mL, at least 2 ⁇ g/mL, at least 5 ⁇ g/mL, at least 6 ⁇ g/mL, at least 10 ⁇ g/mL, at least 15 ⁇ g/mL, at least 20 ⁇ g/mL, at least 25 ⁇ g/mL, at least 50 ⁇ g/mL, at least 100 ⁇ g/mL, at least 125 ⁇ g/mL, at least 150 ⁇ g/mL, at least 175 ⁇ g/mL, at least 200 ⁇ g/mL, at least 225 ⁇ g/mL, at least
  • a subject is administered a dose of an effective amount of one or more antibodies or immunoconjugates of the invention to achieve a serum titer of at least 0.1 ⁇ g/mL, at least 0.5 ⁇ g/mL, at least 1 ⁇ g/mL, at least, 2 ⁇ g/mL, at least 5 ⁇ g/mL, at least 6 ⁇ g/mL, at least 10 ⁇ g/mL, at least 15 ⁇ g/mL, at least 20 ⁇ g/mL, at least 25 ⁇ g/mL, at least 50 ⁇ g/mL, at least 100 ⁇ g/mL, at least 125 ⁇ g/mL, at least 150 ⁇ g/mL, at least 175 ⁇ g/mL, at least 200 ⁇ g/mL, at least 225 ⁇ g/mL, at least 250 ⁇ g/mL, at least 275 ⁇ g/mL, at least 300 ⁇ g/mL, at least 325 ⁇
  • a subject may be administered 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 or more subsequent doses.
  • the invention provides methods of preventing and/or treating a disorder associated with or characterized by aberrant expression and/or activity of GPNMB, said method comprising administering to a subject in need thereof a unit dose of at least O.Olmg/kg, at least 0.1mg/kg, at least 0.2mg/kg, at least 0.4mg/kg, at least 0.6mg/kg, at least 0.8mg/kg, at least lmg/kg, or at least 1.lmg/kg of one or more antibodies or immunoconjugates of the invention.
  • the invention provides methods of preventing and/or treating a disorder associated with or characterized by aberrant expression and/or activity of GPNMB, said method comprising administering to a subject in need thereof a unit dose of at least 0.01 mg/kg, at least 0.1 mg/kg, at least 0.2 mg/kg, at least 0.4 mg/kg, at least 0.6 mg/kg, at least 0.8 mg/kg, at least 1 mg/kg, or at least 1.1 mg/kg of one or more antibodies or immunoconjugates of the invention once every 7 days, preferably, once every 10 days, once every 12 days, once every 14 days, once every 16 days, once every 18 days, once every three weeks, or once a month.
  • an immunoconjuage of the instant invention is administered at a unit dose of about 0.1 mg/kg, about 0.2 mg/kg, about 0.4 mg/kg, about 0.6 mg/kg, about 0.8 mg/kg, about 1.1 mg/kg, or about 1 mg/kg once every 10 to 20 days with 2 to 4 cycles.
  • the present invention provides methods of preventing and/or treating a disorder associated with or characterized by aberrant expression and/or activity of GPNMB, said method comprising: (a) adtninistering to a subject in need thereof one or more doses of a prophylactically or therapeutically effective amount of one or more antibodies or immunoconjugates of the invention; and (b) monitoring the plasma level/concentration of the said administered antibody or antibodies in said subject after administration of a certain number of doses of the said antibody or antibodies.
  • said certain number of doses is 1, 2, 3, 4, 5, 6, 7, or 8 doses of a prophylactically or therapeutically effective amount one or more antibodies or immunoconjugates of the invention.
  • the invention provides a method of preventing and/or treating a disorder associated with or characterized by aberrant expression and/or activity of GPNMB, said method comprising: (a) administering to a subject in need thereof a dose of at least 0.1 mg/kg (preferably at least at least 0.2 mg/kg, at least 0.4 mg/kg, at least 0.6 mg/kg, at least 0.8 mg/kg, at least 1 mg/kg, or at least 1.1 mg/kg) of one or more antibodies or iminunoconjugates of the invention; and (b) administering one or more subsequent doses to said subject when the plasma level of the antibody or antibodies administered in said subject is less than 0.1 ⁇ g/mL, preferably less than 0.25 ⁇ g/mL, less than 0.5 ⁇ g/mL, less than 0.75 ⁇ g/mL, or less than 1 ⁇ g/mL.
  • the invention provides a method of preventing and/or treating a disorder associated with or characterized by aberrant expression and/or activity of GPNMB, said method comprising: (a) administering to a subject in need thereof one or more doses of at least at least 0.1 mg/kg (preferably at least at least 0.2 mg/kg, at least 0.4 mg/kg, at least 0.6 mg/kg, at least 0.8 mg/kg, at least 1 mg/kg, or at least 1.1 mg/kg) of one or more antibodies of the invention; (b) monitoring the plasma level of the administered antibody or antibodies of the invention in said subject after the administration of a certain number of doses; and (c) administering a subsequent dose of the antibody or antibodies of the invention when the plasma level of the administered antibody or antibodies in said subject is less than 0.1 ⁇ g/mL, preferably less than 0.25 ⁇ g/mL, less than 0.5 ⁇ g/mL, less than 0.75 ⁇ g/mL, or less than 1 ⁇ g/mL.
  • Therapies e.g., prophylactic or therapeutic agents
  • other than antibodies or immunoconjugates of the invention which have been or are currently being used to prevent and/or treat a disorder associated with or characterized by aberrant expression and/or activity of GPNMB
  • the dosages of prophylactic or therapeutic agents used in combination therapies of the invention are lower than those which have been or are currently being used to prevent and/or treat a disorder associated with or characterized by aberrant expression and/or activity of GPNMB.
  • the therapies are administered less than 5 minutes apart, less than 30 minutes apart, 1 hour apart, at about 1 hour apart, at about 1 to about 2 hours apart, at about 2 hours to about 3 hours apart, at about 3 hours to about 4 hours apart, at about 4 hours to about 5 hours apart, at about 5 hours to about 6 hours apart, at about 6 hours to about 7 hours apart, at about 7 hours to about 8 hours apart, at about 8 hours to about 9 hours apart, at about 9 hours to about 10 hours apart, at about 10 hours to about 1 1 hours apart, at about 11 hours to about 12 hours apart, at about 12 hours to 18 hours apart, 18 hours to 24 hours apart, 24 hours to 36 hours apart, 36 hours to 48 hours apart, 48 hours to 52 hours apart, 52 hours to 60 hours apart, 60 hours to 72 hours apart, 72 hours to 84 hours apart, 84 hours to 96 hours apart, or 96 hours to 120 hours part.
  • two or more therapies are administered within the same patient visit.
  • one or more antibodies of the invention and one or more other therapies are cyclically administered. Cycling therapy involves the administration of a first therapy (e.g., a first prophylactic or therapeutic agent) for a period of time, followed by the administration of a second therapy (e.g., a second prophylactic or therapeutic agent) for a period of time, optionally, followed by the administration of a third therapy (e.g., prophylactic or therapeutic agent) for a period of time and so forth, and repeating this sequential administration, i.e., the cycle in order to reduce the development of resistance to one of the therapies, to avoid or reduce the side effects of one of the therapies, and/or to improve the efficacy of the therapies.
  • a first therapy e.g., a first prophylactic or therapeutic agent
  • a second therapy e.g., a second prophylactic or therapeutic agent
  • a third therapy e.g., prophylactic or therapeutic agent
  • compositions comprising anti-GPNMB antibodies. Such compositions may be suitable for pharmaceutical use and administration to patients.
  • the compositions typically comprise one or more antibodies of the present invention and a pharmaceutically acceptable excipient.
  • pharmaceutically acceptable excipient includes any and all solvents, dispersion media, coatings, antibacterial agents and antifungal agents, isotonic agents, and absorption delaying agents, and the like, that are compatible with pharmaceutical administration. The use of such media and agents for pharmaceutically active substances is well known in the art.
  • the compositions may also contain other active compounds providing supplemental, additional, or enhanced therapeutic functions.
  • the pharmaceutical compositions may also be included in a container, pack, or dispenser together with instructions for administration.
  • a pharmaceutical composition of the invention is formulated to be compatible with its intended route of administration. Methods to accomplish the administration are known to those of ordinary skill in the art.
  • the administration may, for example, be intravenous, intraperitoneal, intramuscular, intracavity, subcutaneous or transdermal. It may also be possible to obtain compositions which may be topically or orally administered, or which may be capable of transmission across mucous membranes.
  • Solutions or suspensions used for intradermal or subcutaneous application typically include one or more of the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerin, propylene glycol, or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates; and agents for the adjustment of tonicity such as sodium chloride or dextrose.
  • the pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide.
  • compositions suitable for injection include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion.
  • suitable carriers include physiological saline, bacteriostatic water, Cremophor EL (BASF, Parsippany, NJ.) or phosphate buffered saline (PBS).
  • the composition must be sterile and should be fluid to the extent that easy syringability exists. It should be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi.
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyetheylene glycol, and the like), and suitable mixtures thereof.
  • the proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and/or by the use of surfactants.
  • Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent which delays absorption, for example, aluminum monostearate, and gelatin.
  • Oral compositions generally include an inert diluent or an edible carrier. They can be enclosed in gelatin capsules or compressed into tablets. For oral administration, the antibodies can be combined with excipients and used in the form of tablets, troches, or capsules. Pharmaceutically compatible binding agents, and/or adjuvant materials can be included as part of the composition.
  • the tablets, pills, capsules, troches, and the like can contain any of the following ingredients, or compounds of a similar nature; a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or com starch; a lubricant such as magnesium stearate or Sterotes; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring.
  • a binder such as microcrystalline cellulose, gum tragacanth or gelatin
  • an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or com starch
  • a lubricant such as magnesium stearate or Sterotes
  • a glidant such as colloidal silicon dioxide
  • Systemic administration can also be by transmucosal or transdermal means.
  • penetrants appropriate to the harrier to be permeated are used in the formulation.
  • penetrants are generally known in the art, and include, for example, detergents, bile salts, and fusidic acid derivatives.
  • Transmucosal administration may be accomplished, for example, through the use of lozenges, nasal sprays, inhalers, or suppositories.
  • compositions may be capable of transmission across mucous membranes in intestine, mouth, or lungs (e.g., via the FcRn receptor-mediated pathway as described in U.S. Pat.
  • the active compounds may be formulated into ointments, salves, gels, or creams as generally known in the art.
  • the antibodies may be delivered in the form of an aerosol spray from pressured container or dispenser, which contains a suitable propellant, e.g., a gas such as carbon dioxide, or a nebulizer.
  • a suitable propellant e.g., a gas such as carbon dioxide, or a nebulizer.
  • the presently disclosed antibodies are prepared with carriers that will protect the compound against rapid elimination from the body, such as a controlled release formulation, including implants and microencapsulated delivery systems.
  • Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Methods for preparation of such formulations will be apparent to those skilled in the art.
  • Liposomal suspensions containing the presently disclosed antibodies can also be used as pharmaceutically acceptable carriers. These can be prepared according to methods known to those skilled in the art, for example, as described in U.S. Pat. No. 4,522,811.
  • dosage unit form refers to physically discrete units suited as unitary dosages for the subject to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • Toxicity and therapeutic efficacy of the composition of the invention can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD 5O (the dose lethal to 50% of the population) and the ED 50 (the dose therapeutically effective in 50% of the population).
  • the dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD 50 /ED 50 .
  • Compositions that exhibit large therapeutic indices are preferred.
  • the therapeutically effective dose can be estimated initially from cell culture assays.
  • suitable bioassays include DNA replication assays, clonogenic assays and other assays as, for example, described in the Examples.
  • the data obtained from the cell culture assays and animal studies can be used in formulating a range of dosage for use in humans.
  • a dose may be formulated in animal models to achieve a circulating plasma concentration range that includes the IC50 (i.e., the concentration of the antibody which achieves a half-maximal inhibition of symptoms). Circulating levels in plasma may be measured, for example, by high performance liquid chromatography.
  • the effects of any particular dosage can be monitored by a suitable bioassay.
  • the dosage lies preferably within a range of circulating concentrations with little or no toxicity. The dosage may vary depending upon the dosage form employed and the route of administration utilized.
  • Antibodies can be modified to become immunotoxins utilizing techniques that are well known in the art (Vitetta 1993, Immunol Today 14:252; U.S. Patent No. 5,194,594).
  • Cyotoxic immunoconjugates are known in the art and have been used as therapeutic agents. Such immunoconjugates may for example, use maytansinoids (US 6,441,163), tubulin polymerization inhibitor, auristatin (Mohammad et al, 1999 Int. J. Oncol 15(2):367-72; Dorot ⁇ na et al, 2003 Nature Biotechnology 21(7): 778-784), dolastatin derivatives (Ogawa et al, 2001 Toxicol Lett. 121(2):97-106) 21(3)778- 784), Mylotarg® (Wyeth Laboratories, Philidelphia, PA); maytansinoids (DMl), taxane or mertansine (ImmunoGen Inc.).
  • Immunoradiopharmaceuticals utilizing anti-GPNMB antibodies may be prepared utilizing techniques mat are well known in the art (Junghans et al. in Cancer Chemotherapy and Bioth ⁇ rapy 655-686 (2d edition, Chafner and Longo, eds., Lippincott Raven (1996);US. Patent Nos. 4,681,581, 4, 735,210, 5,101,827, 5,102,990 (RE 35,500), 5,648,471, and 5,697,902). Each of the immunotoxins and radiolabeled antibody molecules selectively kill cells expressing GPNMB. Radiolabels are known in the art and have been used for diagnostic or therapeutic radioimmuno conjugates.
  • radiolabels include, but are not limited to, the following: radioisotopes or radionuclides (e.g., 3 H, 14 C, 15 N, 35 S, 90 Y, 99 Tc, 111 In, 125 I, 131 I, 177 Lu, 105 Rh, Rhenium-186, Rhenium-188, Samarium- 153, Copper-64, Scandium-47).
  • radioisotopes or radionuclides e.g., 3 H, 14 C, 15 N, 35 S, 90 Y, 99 Tc, 111 In, 125 I, 131 I, 177 Lu, 105 Rh, Rhenium-186, Rhenium-188, Samarium- 153, Copper-64, Scandium-47).
  • radionuclides which have been used in radioimmunoconjugate guided clinical diagnosis include, but are not limited to: 131 I, 125 1, 123 j ⁇ 99 ⁇ 67 Q a ⁇ ag we jj ag i n j n Antibodies have also been labeled with a variety of radionuclides for potential use in targeted immunotherapy (see Peirersz et ah, 1987). These radionuclides include, for example, ' * Re and ' Re as well as 90 Y, and to a lesser extent ' " Au and bl Cu. I-( 131 ) ⁇ see for example U. S. Pat. No. 5, 460, 785) . Radio therapeutic chelators and chelator conjugates are known in the art (U.S. 4,831,175, 5,099,069, 5,246,692, 5,286,850, and 5,124,471).
  • Recombinant human GPNMB (SEQ ID NO:289), specifically the extra-cellular domain (ECD) was prepared for use as the immunogen.
  • ECD extra-cellular domain
  • cDNA encoding the ECD of GPNMB with a C-terminus V5-HIS tag was transfected into HEK 293 cells, expressed and purified using cation exchange chromatography with a POROS HS 50 (Applied Biosystems, Foster City, CA).
  • Sample was eluted with IM NaCl at a pH of 5.5, followed by metal affinity chromatography (Pharmacia metal chelate 5 mL). The sample was eluted against a linear gradient from 10-500 mM imidazole over 10 CV (column volumn). Dialysis occurred using 2OmM Tris/ 5OmM NaCl at pH 7.4 (2L x 2). The sample was then filtered through a 0.22 ⁇ m filter.
  • Example 2 Immunization A preferred method for generating fully human antibodies uses XenoMouse ⁇ strains of mice which have been engineered to contain 245 kb and 190 kb-sized germline configuration fragments of the human heavy chain locus and kappa light chain locus (Green et al. 1994 Nature Genetics 7:13-21; Mendez et al. 1997 Nature Genetics 15:146-156; Green and Jakobovits, 1998 J. Exp. Med. 188:483-495; U.S. Patent Nos.
  • one or more V H genes, one or more D H genes, one or more J H genes, a mu constant region, and a second constant region are formed into a construct for insertion into an animal (Taylor et al, 1992, Chen et al, 1993, Tuaillon et al, 1993, Choi et al, 1993, Lonherg et al, (1994), Taylor et al, (1994), and Tuaillon et al., (1995), Fishwild et al, (1996); U.S. Patent Nos.
  • ⁇ K XenoMouse® may be used to generate anti-GPNMB antibodies utilizing lambda V regions. Such antibodies are within the scope of the invention.
  • Immunization GPNMB-V5His immunogen (as prepared in Example 1) was used as an antigen.
  • XenoMouse ® mice Monoclonal antibodies against GPNMB were developed by sequentially immunizing XenoMouse ® mice (XenoMouse' XMG2 strain), Abgenix, Inc. Fremont, CA. XenoMouse ® animals were immunized via footpad route for all injections. The total volume of each injection was 50 ⁇ l per mouse, 25 ⁇ l per footpad. For cohort 1 (10 XMG2 mice), the initial immunization was with 10 ⁇ g of
  • the subsequent five boosts were made with 5 ⁇ g of GPNMB-V5His admixed 1:1 (v/v) with 100 ⁇ g alum gel in pyrogen-free D-PBS.
  • the seventh boost consisted of 5 ⁇ g of GPNMB-V5His admixed 1:1 (v/v) with TITERMAX GOLD®(Sigma; cat.
  • the eighth injection consisted of 5 ⁇ g of GPNMB -V5His admixed 1 :1 v/v with 100 ⁇ g alum gel .
  • a final boost was made with 5 ⁇ g GPNMB-V5His in pyrogen-free DPBS, without adjuvant.
  • the XenoMouse ® mice were immunized on days 0, 3, 6, 10, 14, 17, 23, and 27 for this protocol and fusions were performed on day 31. The bleed was made through Retro-Orbital Bleed procedure on day 21 after the sixth boost .
  • cohort 2 (10 XMG2 mice), the initial immunization was with 10 ⁇ g of
  • GPNMB-V5His admixed 1:1 (v/v) with 100 ⁇ g alum gel per mouse.
  • the subsequent two boosts were made with 5 ⁇ g of GPNMB-V5His admixed 1 :1 (v/v) with 100 ⁇ g alum gel in pyrogen-free D-PBS.
  • the fourth boost consisted of 5 ⁇ g of GPNMB-V5His admixed 1:1 (v/v) with TITERMAX GOLD®(Sigma; cat. # T2684).
  • the following fifth to seventh injection consisted of 5 ⁇ g of GPNMB-V5His admixed 1:1 v/v with 100 ⁇ g alum gel .
  • the eighth injection and final boost was made with 5 ⁇ g GPNMB-V5His in pyrogen-free
  • mice were immunized on days 0, 3, 7, 11, 14,
  • the footpad injection was performed by the following protocol using only the ventral surface of both hind limb paws.
  • a solution was injected beneath the skin without piercing the muscle tissue by using an insulin 1/2 mL syringe with attached 28 or 30 gauge x 1/2" needle.
  • the mouse to be injected was grasped by the loose fur along its neck and back so that it was immobilized and was turned over so the ventral side was accessible.
  • the hind limb of the mouse was grasped and the needle was inserted (bevel side up) at the ankle, threading just under the skin until the needle tip reached the paw.
  • the needle was inserted along the outside length of the hind foot carefully, to avoid the vein located towards the inner side of the foot. Once the tip of the needle reached the paw, the solution was injected slowly until resistance was felt or the designated volume had been dispensed. The needle was then withdrawn and the second hind foot injected in the same manner.
  • Table 4 provides the immunization schedule for the 2 groups of mice.
  • Anti- GPNMB antibody titers in the serum from immunized XenoMouse ® mice were determined by ELISA. Briefly, three sets of ELISAs were set up. GPNMB (+NMB) at 1 ⁇ g/mL, GPNMB(-NMB) at 1 ⁇ g/mL, and NMB at 1 ⁇ g/mL were coated onto Costar Labcoat Universal Binding Polystyrene 96-well plates (Corning, Acton, MA) overnight at 4 0 C in Antigen Coating Buffer (0.1 M Carbonate Buffer, pH 9.6 NaHCO 3 (MW 84) 8.4 g/L).
  • Antigen Coating Buffer 0.1 M Carbonate Buffer, pH 9.6 NaHCO 3 (MW 84) 8.4 g/L).
  • Hybridoma cell lines were generated from immunized mice demonstrated to have anti-GPNMB titers using standard techniques (see Mendez et al, 1997, Nat Genet. 15: 146- 156).
  • lymph nodes were harvested and pooled from each cohort.
  • the lymphoid cells were dissociated by grinding in
  • DMEM to release the cells from the tissues, and the cells were suspended in DMEM.
  • the cells were counted, and 0.9 mL DMEM per 100 million lymphocytes was added to the cell pellet to resuspend the cells gently but completely.
  • the cells were labeled by incubating the cells with the magnetic beads at 4°C for 15 minutes.
  • the magnetically-labeled cell suspension containing up to 10 8 positive cells (or up to 2x10 9 total cells) was loaded onto a LS+ column and the column washed with DMEM. The total effluent was collected as the CD90-negative fraction (most of these cells were expected to be B cells).
  • the fusion was performed by mixing washed enriched B cells from above and nonsecretory myeloma P3X63Ag8.653 cells purchased from ATCC, cat.# CRL 1580
  • Electro-cell fusion was performed using a fusion generator, model ECM2001, Genetronic, Inc., San Diego, CA.
  • the fusion chamber size used was 2.0 mL, using the Abgenix, Inc. optimum instrument settings to do ECF.
  • ECF the cell suspensions were carefully removed from the fusion chamber under sterile conditions and transferred into a sterile tube containing the same volume of Hybridoma Culture Medium (DMEM (JRH Biosciences), 15% FBS (Hyclone), supplemented with L-glutamine, pen/strep, OPI (oxaloacetate, pyruvate, bovine insulin) (all from Sigma) and IL-6 (Boehringer Mannheim)).
  • DMEM Hybridoma Culture Medium
  • FBS Hybridoma Culture Medium
  • OPI oxaloacetate, pyruvate, bovine insulin
  • Hybridoma Selection Medium Hybridoma Culture Medium supplemented with 0.5x HA (Sigma, cat. # A9666)
  • Hybridoma Selection Medium 0.5x HA (Sigma, cat. # A9666)
  • the cells were mixed gently and pipetted into 96-well plates and allowed to grow. On day 7 or 10, one-half the medium was removed, and the cells were re-fed with Hybridoma Selection Medium.
  • the ELISA plates (Fisher, Cat. No. 12-565-136) were coated with 50 ⁇ L/well of GPNMB (1 ⁇ g/mL) in Coating Buffer (0.1 M Carbonate Buffer, pH 9.6, NaHCO 3 8.4 g/L), then incubated at 4 0 C overnight. After incubation, the plates were washed with Washing Buffer (0.05% Tween 20 in PBS) three times.
  • Coating Buffer 0.1 M Carbonate Buffer, pH 9.6, NaHCO 3 8.4 g/L
  • Blocking Buffer (0.5% BSA, 0.1% Tween 20, 0.01% Thimerosal in Ix PBS) were added and the plates were incubated at room temperature for 1 h. After incubation, the plates were washed with Washing Buffer three times. Aliquots (50 ⁇ L/well) of hybridoma supernatants and positive and negative controls were added, and the plates were incubated at room temperature for 2 h.
  • the positive control used throughout was serum from the relevant GPNMB immunized XenoMouse ® mouse and the negative control was serum from the KLH-immunized relevant strain of XenoMouse ® mouse. After incubation, the plates were washed three times with Washing Buffer.
  • the old culture supernatants from the positive hybridoma cells growth wells based on primary screen were removed completely and the IL-Ib positive hybridoma cells were suspended with fresh hybridoma culture medium and were transferred to 24-well plates. After 2 days in culture, these supernatants were ready for a secondary confirmation screen.
  • the positives in the first screening were screened in GPNMB binding ELISA described as above, and two sets of detective system for the secondary confirmation ELISA, one set for hlgG detection, one set for human Ig kappa light chain detection (goat anti-hlg kappa-HRP, Southern Biotechnology, Cat. No. 2060-05) in order to demonstrate fully human composition for both heavy and light chains.
  • the two sets of ELISA procedures were identical to the descriptions above except the three different detection antibodies were used separately. All positive hits from the secondary confirmation ELISA assay were counter screened for binding to immunogen. by ELISA in order to exclude those that cross-react with IL-Ia.
  • the ELISA plates (Fisher, Cat. No. 12-565-136) were coated with 50 ⁇ L/well of irrelevant V5His-fusion protein, lug/mL in Coating Buffer (0.1 M Carbonate Buffer, pH 9.6, NaHCO 3 8.4 g/L), then incubated at 4°C overnight. The remaining procedures were identical to the descriptions above. There are 33 fully human GPNMB specific monoclonal antibodies that were generated. Hybridoma supernatants were screened for binding to GPNMB by ELISA as described above in Example 2. Results are shown in Table 6. Table 6. Hybridoma anti-GPNMB activity.
  • Hybridoma cell supernatants (29) were analyzed for binding to GPNMB by BiaCore® 2000 biosensor equipped with a research-grade CM5 sensor chip.
  • a 1 :25 dilution of cell supernatant was passed over a protein A surface for 5 min followed by washing the surface for 10 mins.
  • GPNMB was injected for 90 sec. over the surface at a concentration of 880 nM followed by dissociation.
  • Double-referenced binding data were obtained by subtracting the signal from a control flow cell and subtracting the baseline drift of a buffer injected just prior to the antigen injection.
  • GPNMB binding ddata for each niAb was normalized for the amount of niAb captured on each surface.
  • MxIiIgG conjugated beads are prepared for coupling to primary antibody.
  • each bead is added to each well of filter plate, then washed once by adding 100 ⁇ L/well wash buffer and aspirating.
  • Antigen and controls are added to filter plate 50uL/well then covered and allowed to incubate in the dark for 1 hour on shaker.
  • a secondary unknown antibody is added at 50 ⁇ L/well using the same dilution (or concentration if known) as is used for the primary antibody. The plates are then incubated in the dark for 2 hours at RT on shaker followed by a wash step.
  • Bin 1 included GPNMB antibodies (1.2.1), (1.10.1), and (2.22.1).
  • Bin 2 included GPNMB antibodies (2.3.1) and (1.15.1), and
  • Bin 3 included GPNMB antibody (2.10.1).
  • the results of the binning assays are provided below in Tables 8 and 9.
  • Example 5 GPNMB Immuno histochemistry (IHC) Analysis Anti-GPNMB monoclonal antibodies were evaluated for reactivity with frozen and fixed tissue specimens. Tissue sections (5 ⁇ m) were cut from formalin fixed and paraffin embedded tissue samples and were rehydrated through incubations in xylene and a graded ethanol series terminating in PBS. Endogenous peroxidase activity was quenched in a 3% solution of hydrogen peroxide in methanol. Tissue sections were blocked in blocking buffer (5% BSA (Sigma), 1% goat serum
  • Sections were washed in 3 changes of PBS for 5 to 10 minutes each and incubated with a 1 :200 dilution of streptavidin conjugated horseradish peroxidase (Jackson Immunolabs) in blocking buffer for 30 minutes and then washed as before. Antibody was detected using DAB reagent (Vector labs). Sections were counterstained in hematoxylin (Fisher Scientific) and dehydrated through alcohol and xylene and coverslipped with permount (Fisher Scientific).
  • Anti-GPNMB Mabs 2.22.1 and 2.22.2 were used to stain normal and tumor human tissue microarrays (IMPATH, Los Angeles, CA). Positive staining was seen in lung, ovarian, renal, esophagus, and head & neck carcinomas, squamous cell carcinoma, melanomas and normal skin specimens. Melanoma and lung carcinomas showed the highest staining intensities with subcellular staining located in the membrane and cytoplasm. Anti-GPNMB Mab 2.10.2 also stained primary melanoma.
  • Anti-GPNMB antibody staining of melanoma tissue microarray showed a large proportion of melanoma cases to be positively stained as shown in Tables 10 and 11.
  • SCC lung squamous cell carcinoma
  • the specificity of anti-GPNMB antibodies to cell membrane-bound GPNMB protein expressed by melanoma cancer cell line, UACC-62 was analyzed by FACS analysis.
  • a renal cancer cell line, TKlO, which does not express GPNMB antigen was used as a negative control.
  • Isotype matched antibody pK16.3 was used as a negative control.
  • Cells were washed twice with PBS (Ca and Mg free), incubated with Versene at 37 0 C until cells detached, counted and aliquoted at 1 million cells per assay tube. Cells were then washed twice and resuspended in ice-cold FACS buffer (0.01M HEPES, 0.15M NaCl, 0.1% NaN 3 and 4% FBS).
  • Example 7 FACS analysis of anti-GPNMB MAb binding to Lymphoma and Leukemia
  • GPNMB GPNMB on the surface of hematopoietic malignant cells
  • cell lines derived from various lymphomas and leukemias were incubated with anti-GPNMB antibody and analyzed by FACS. Lymphoma or leukemia derived cells were washed twice with ice-cold FACS buffer and resuspended at 1 million cells per assay tube. MAb 1.15.1 antibody at 1 ⁇ g/mL was added to cells and cells were incubated on ice for 30 min. Cells were then washed 2-3 times and resuspended in 1 ml_ of ice-cold FACS buffer.
  • UACC-62, a GPNMB antigen expressing cell line, and TKlO, a non-expressing cell line were plated onto flat bottom 96-well tissue culture plates (Becton Dickinson, Franklin Lakes, NJ, USA) at a density of 3000 cells per well. Once the cells reached -25% confluency, 100 ng/well of secondary antibody-toxin conjugate (goat anti -human IgG- saporin; Advanced Targeting Systems, San Diego, USA, HUM-ZAP; cat. # IT-22) was added.
  • Anti-GPNMB MAbs 2.10.2, 2.22.1, 1.15.1 or isotype control niAb (pK16.3) were added to each well at a final concentration of 10 or 50 ng/mL.
  • An anti-EGFR monoclonal antibody (MS-269-PABX, NeoMarkers, Fremont, CA, USA) was used as a positive primary antibody control.
  • Chemotherapy reagent 5-FU at 600 uM was used as a positive reagent control.
  • the cells were trypsinized, transferred to 6-well tissue culture plates and incubated at 37 0 C. Plates were examined daily and between 8-10 days, all plates were Giemsa stained and colonies were counted.
  • Example 10 Cell killing by Auristatin-E (AE) Conjugated anti-GPNMB Antibodies UACC-62 and TKlO cells were plated onto flat bottom 96-well tissue culture plates
  • IC 50 of 1.15.1 -AE mediated killing on GPNMB positive and negative cells are presented in Table 15. Unconjugated 1.15.1 and AE-conjugated 2.6.2 had no effect on growth of all the melanoma cell lines tested. However, cell lines SK-Mel2, WM-266-4, G361, UACC-257, UACC-62, RPMI-7951 and SK-Mel5 were susceptible to 1.15.1-AE mediated killing in a dose-dependent fashion. SK-Mel2 demonstrated the lowest IC5 0 in this study (Table 15). These results show the specific and cytotoxic effects of AE conjugated 1.15.1 on most of GPNMB expressing melanoma cells.
  • Lymphoma or leukemia cell lines were mixed with methylcellulose base media (R&D Systems, USA) and in various concentrations of unconjugated and Auristatin E- co ⁇ jugated 1.15.1 antibody before plating onto 6-well tissue culture plates (Becton Dickinson, Franklin Lakes, NJ, USA).
  • MAb 2.6.2-AE was also included as a conjugated isotype control in this study because it does not bind to GPNMB expressing cells. Plates were incubated at 37 0 C and examined daily. On days 14-18, colonies on the plates were counted.
  • the IC 50 of 1.15.1-AE induced cell killing on antigen expressing cells is presented in Table 16.
  • Example 13 CROl l-vcMMAE Inhibits the Growth of Human SK-MEL-2 Melanoma Xenografts Leading to Complete Regression of Established Melanoma Tumo ]n Athymic Mice (Study N-386) Study N-386 was performed to assess the potency and therapeutic efficacy of the antibody-drug conjugate, CROl l-vcMMAE, against the established human SK-MEL-2 melanoma xenograft in athymic mice.
  • Materials and Methods Test Animals: Five- to 6-week old athymic mice (CD-I nu/nu females), used for human tumor xenografts, were obtained from Harlan Laboratories (Indianapolis, IN).
  • the SK-MEL-2 human melanoma (ATCC #HTB-68) was derived from a metastatic site (skin of thigh) of a 60 year old Caucasian male with malignant melanoma, and the SK- MEL-5 human melanoma (ATCC #HTB-70) was derived from a metastatic site (axillary lymph node) of a 24 year old Caucasian female with malignant melanoma (see Fogh et al, J. Natl. Cancer Inst. 59: 221-226 (1977)). Both cell lines were obtained from the American Type Culture Collection.
  • tumor size (in mg) was calculated using a standard formula, (W x L)/2, assuming a specific gravity of 1.0. Tumor size and body weights were assessed twice weekly. Mice were examined daily, however, and moribund animals were humanely euthanized if clinical indications of excessive pain or distress were noted ⁇ i.e.,
  • CROl l-vcMMAE 0.625 - 20 mg/kg i.v., every 4 days for a total of 4 treatments (i.e., q4d X4); saline and phosphate-buffered saline controls (i.v., q4d X4); and two known anti-tumor reference agents, vinblastine sulfate (i.v., 1.7 mg/kg, q4d X4) and paclitaxel (i.v., 24 mg/kg, q2d X4).
  • the reference agents were administered at the maximum tolerated dose (MTD) determined in prior studies.
  • SK-MEL-2 tumors have a high "take” rate in immunocompromised hosts (97 %) and a low rate of spontaneous regression (3 %) (Dykes et al, Development of human tumor xenograft models for in vivo evaluation of new antitumor drugs, in
  • Vinblastine produced a very slight, but not significant, anti-tumor effect (P ⁇ 0.20); in this and other tumor models (e.g., SK-MEL-5) vinblastine produces noticeable tumor growth inhibition, but which is only occasionally significant.
  • Paclitaxel showed significant tumor growth inhibition and tumor stasis (i.e., 100% growth inhibition) for approximately 2 weeks after treatment commenced (P ⁇ 0.0077).
  • CRO 11 -vcMMAE produces substantial, dose-dependent and reproducible anti-tumor effects that begin as tumor growth inhibition but soon lead to complete regression of established human melanoma xenografts; the regressions are long-lived and re-growth of tumors after successful therapy has not been observed.
  • variable heavy chains and the variable light chains for the antibodies shown in Table 17 were sequenced to determine their DNA and protein sequences.
  • GIPDRFSGSGSGTDFTLTISRLEPEDFAVYYC QQYGSSIT FGQGTRLEIKR 3' (SEQ ID NO:11)
  • CTCCTCAGCC 3' (SEQ ID NO:19)
  • TCCTCAGCC 3' (SEQ ID NO:37)
  • GIPARFSGSGSGTEFTLTISSLQSEDFAVYYC QQYHYWPT FGPGTKVDIKR 3' (SEQ ID NO:119)
  • Example 16 Use of anti-GPNMB Antibodies as a Diagnostic Agent Detection of GPNMB antigen in a sample:
  • ELISA Enzyme-Linked Immunosorbent Assay
  • wells of a microtiter plate such as a 96-well microtiter plate or a 384-well microtiter plate, are adsorbed for several hours with a first fully human monoclonal antibody directed against GPNMB.
  • the immobilized antibody serves as a capture antibody for any of the GPNMB that may be present in a test sample.
  • the wells are rinsed and treated with a blocking agent such as milk protein or albumin to prevent nonspecific adsorption of the analyte.
  • test sample suspected of containing the GPNMB antigen, or with a solution containing a standard amount of GPNMB antigen.
  • a sample may be, for example, a serum sample from a subject suspected of having levels of circulating GPNMB considered to be diagnostic of a pathology.
  • the wells After rinsing away the test sample or standard, the wells are treated with a second fully human monoclonal anti-GPNMB antibody that is labeled by conjugation with biotin.
  • the labeled anti-GPNMB antibody serves as a detecting antibody.
  • the wells After rinsing away excess second antibody, the wells are treated with avidin-conjugated horseradish peroxidase (HRP) and a suitable chromogenic substrate.
  • HRP horseradish peroxidase
  • the concentration of the antigen in the test samples is determined by comparison with a standard curve developed from the standard samples.
  • This ELISA assay provides a highly specific and very sensitive assay for the detection of the GPNMB antigen in a test sample. Determination of GPNMB antigen concentration in patients:
  • a sandwich ELISA can also be used to quantify GPNMB levels in human serum.
  • the 2 fully human monoclonal anti-GPNMB antibodies used in the sandwich ELISA recognize different epitopes on the GPNMB molecule.
  • the ELISA is performed as follows: 50 ⁇ l of capture anti-GPNMB antibody in coating buffer (0.1 M NaHC ⁇ 3, pH 9.6) at a concentration of 2 ⁇ g/mL is coated on ELISA plates (Fisher). After incubation at 4°C overnight, the plates are treated with 200 ⁇ l of blocking buffer (0.5% BSA, 0.1% Tween 20, 0.01% Thimerosal in PBS) for 1 hr at 25°C.
  • the plates are washed (3x) using 0.05% Tween 20 in PBS (washing buffer, WB). Normal or patient sera (Clinomics, Bioreclaimation) are diluted in blocking buffer containing 50% human serum. The plates are incubated with serum samples overnight at 4°C, washed with WB, and then incubated with 100 ⁇ l/well of biotinylated detection anti-GPNMB antibody for 1 hr at 25 0 C. After washing, the plates are incubated with HRP-Streptavidin for 15 min, washed as before, and then treated with 100 ⁇ l/well of o-phenylenediamine in H2O2 (Sigma developing solution) for color generation.
  • PBS washing buffer, WB
  • Normal or patient sera are diluted in blocking buffer containing 50% human serum.
  • the plates are incubated with serum samples overnight at 4°C, washed with WB, and then incubated with 100 ⁇ l/well of biotinylated detection anti-GPNMB antibody for
  • a cancer in a subject based on expression levels of the GPNMB antigen.
  • samples of blood are taken from subjects diagnosed as being at various stages in the progression of the disease, and/or at various points in the therapeutic treatment of the cancer.
  • concentration of the GPNMB antigen present in the blood samples is determined using a method that specifically determines the amount of the antigen that is present. Such a method includes an ELISA method, such as the method described in the previous diagnostic examples.
  • a range of concentrations of the antigen that may be considered characteristic of each stage is designated.
  • a sample of blood is taken from the subject and the concentration of the GPNMB antigen present in the sample is determined.
  • the concentration so obtained is used to identify in which range of concentrations the value falls.
  • the range so identified correlates with a stage of progression or a stage of therapy identified in the various populations of diagnosed subjects, thereby providing a stage in the subject under study.
  • a subject suspected of having an ovarian cancer tumor is identified and a tissue sample from the suspected tumor is removed for testing.
  • the removed tissue is then contacted with anti-GPNMB antibodies having a colorimetric label.
  • a determination is made of whether the anti-GPNMB antibodies bind specifically to the removed tissue. Binding is indicative of cancereous tissue while the absense of binding is indicative of noncancerous tissue.
  • the patient's conditition is diagnosed accordingly to facilitate subsequent testing, counseling, and/or treatment.
  • Example 18 Treating Cancer With Antibodies against GPNMB
  • Targeting GPNMB on tumor cells is useful to treat a subject at risk for or afflicted with cancer. Such a subject would benefit from treatment with an anti-GPNMB antibody of the present invention.
  • antibodies are administered in an outpatient setting by weekly administration at about 0.1-1.0 mg/kg dose by slow intravenous (IV) infusion.
  • the appropriate therapeutically effective dose of an antibody is selected by a treating clinician and would range approximately from 1 iig/kg to 20mg /kg, from 1 ⁇ _g/kg to 10 mg/kg, from 1 ⁇ .g/kg to 1 mg/kg, from 10 ⁇ g/kg to 1 mg/kg, from 10 j ⁇ g/kg to 100 ⁇ g/kg, from 100 jxg/kg to 1 mg/kg, and from 500 j ⁇ g/kg to 5 mg/kg.
  • the antibodies are also used to prevent and/or to reduce severity and/or symptoms of disease associated with GPNMB-related disorders.
  • Treatment groups include a group not receiving antibody treatment and groups treated with different doses of anti-GPNMB antibody. Individuals are followed prospectively and individuals receiving antibody treatment exhibit an improvement in their condition.
  • mice were implanted by trocar with fragments of SK-ME-2 melanoma and, after the tumors became established, treatment with CROl l-vcMMAE and various components was tested to demonstrate the specificity of anti-tumor effects of this agent.
  • Control groups dosed with either the phosphate-buffered saline (vehicle) or the excipients of the immunoconjugate preparation (3% DMSO, sucrose, phosphate medium) steadily increased in tumor size to a maximum of 2,000 mg, at which time they were removed from the study. No apparent or statistically significant anti-tumor effects were observed.
  • CROl l- vcMMAE treatment at 5 mg/kg/treatment, q4d x4) produced measurable inhibition after the first 2 doses. Tumor growth inhibition continued until no discernible tumor was detected in all 6 of the test animals ( Figure 4). In preliminary studies, tumor regression was complete and was not followed by regrowth of the tumor despite lengthy observation periods (up to 200 days).
  • the regressions produced by the immunoconjugate were not due to the individual components of the immunoconjugate nor to components of the formulation of that immunoconjugate. This is demonstrated by the lack of tumor growth inhibition after treatment with CROI l antibody alone (group 3) or free monomethylauristatin E (group 4), where the doses applied were identical to that contained in the intact immunoconjugate. Furthermore, the lack of anti-tumor effects noted with free MMAE suggests that anti-tumor effects from MMAE as a result of slow release from the antibody-drug conjugate may not explain the anti-tumor effects of the immunoconjugate.
  • Example 20 CROll-vcMMAE Inhibits the Growth of Human SK-MEL-5 Melanoma Xenografts Leading to Complete Regression of Established Melanoma Tumors in Athymic Mice (CROl l-ONC-3)
  • CROl l- vcMMAE produced tumor growth delay leading to complete regressions of established SK- MEL-5 melanoma xenografts (see tabular insert to Figure 5 for proportions of animals with complete regressions). Complete regressions occurred at CROl l-vcMMAE doses of 2.5 mg/kg/treatment, but not at 1.25 mg/kg/treatment.
  • CROl l-vcMMAE exerts substantial, dose-dependent anti-tumor effects against established xenografts of the SK-MEL-5 human melanoma. After just one or two treatments significant tumor growth inhibition is noted and which leads to long-term tumor-free survivors. Complete regressions occurred at doses of > 2.5 mg/kg i.v., q4d X4.
  • Example 21 Pharmacokinetics of CROll-vcMMAE (CROl 1-PK-lA)
  • CROl 1 antibody component of CRO 11 -vcMMAE was measured by a sandwich style enzyme-linked immunosorbent assay (ELISA) where serum was added to the wells of microtiter plates coated with the cognate antigen (GPNMB, CG56972-03) for the CROI l antibody, and the amount of human antibody were detected with an anti-globulin conjugated to the signal generator (horseradish peroxidase).
  • ELISA enzyme-linked immunosorbent assay
  • A Pre-exponential constant for alpha phase
  • Alpha Exponential rate constant for alpha phase
  • AUC Total area under the curve from 0 to infinity
  • B Pre- exponential constant for beta phase
  • Beta Exponential rate constant for beta phase
  • Cl Total or systemic clearance
  • C maX Maximum observed concentration
  • MRT Mean residence time
  • Volume Volume of central compartment
  • Vss Steady-state volume of distribution.
  • the CROl 1-vcMMAE antibody-drug conjugate has a serum-concentration profile which favors continuous exposure sufficient for disruption and eradication of melanoma xenografts.
  • the durability of CROl l-vcMMAE in vivo ⁇ e.g., athymic mice) is comparable to other Auristatin E immunoconjugates.
  • GPNMB was recently shown to be expressed in glioblastoma and to mediate the in vitro and in vivo invasiveness of glioblastoma-derived tumor cells (see,e.g., Loging et al, Genome Res. 10:1393-1402 (2000); and Rich et al, J. Biol. Chem. 278:15951-15975 (2003)). To confirm and extend these findings to additional cancer types, we examined the expression of GPNMB transcripts in human cancer cell lines and tissues.
  • RTQ-PCR analysis was performed with an ABI Prism 7700 Sequence Detection System using TaqMan reagents (PE Applied Biosystems, Foster City, CA). Equal quantities of normalized RNA's were used as a template in PCR reactions for 40 cycles with GPNMB- specific primers to obtain threshold cycle (C T ) values. The following primers (5'-3') were used:
  • Flow cytometry Quantitative analysis of GPNMB expression on the cell surface of cell lines was determined by flow cytometry. Approximately 1 x 10 6 cells were harvested, washed and incubated with a saturating amount (10 ⁇ g/mL) of either CROl 1 or isotype- matched control antibody in staining buffer containing PBS (pH 7.4), 4% FBS and 0.1% NaN3 for 30 min on ice, followed by washing and staining with R-Phycoerythrin (PE)- conjugated goat-anti-human antibody (Jackson ImmunoResearch Laboratories, Inc, West Grove, PA) at 1 : 100 for 30 min on ice.
  • PE Physical Coerythrin
  • Immunoprecipitation and immnnoblot analysis Cells were harvested and lysed on ice for 30 min in lysis buffer containing 1 % NP-40, 0.15 M NaCl, 0.02 M Tris-HCl, 10% glycerol, 0.01 M EDTA and complete protease inhibitor mixture (Roche Molecular Biochemicals, Indianapolis, IN). Supernatants were collected and the protein concentration was determined with the BCA Protein Assay Kit (Pierce, Rockford, IL).
  • Clonogenic Assays The growth-inhibitory activity of CROl 1-vcMMAE was determined by clonogenic assay. Cells were plated in 96-well plates and allowed to recover overnight. Unconj ugated CRO 11 , free MMAE, CRO 11 -vcMMAE or isotype-matched vcMMAE conjugated antibody at various concentrations was added to sub-confluent cell cultures and incubated for 4 days at 37°C. The cells were then transferred into 6-well plates and allowed to form colonies. Colonies were stained with Giemsa stain (Sigma) and counted. The surviving cell fractions were calculated based upon the ratio of the treated sample and the untreated control. The results were expressed as a percentage of control using GraphPad Prism Version 4 software. The IC50 was defined as the concentration resulting in a 50% reduction of colony formation compared to untreated control cultures.
  • CRO 11 -vcMMAE The cell cycle effects of CRO 11 -vcMMAE were evaluated after treating cells in complete growth medium for 24 or 48 hr. Briefly, cells were pulsed at the indicated times with 30 ⁇ M of bromodeoxyuridine (BrdU, Sigma) for 30 min, harvested, fixed and permeabilized in methanol. Nascent DNA synthesis was detected by anti- bromodeoxyuridine-FITC (BD Biosciences, San Jose, CA) staining. Total DNA content was detected using propidium iodide (PI, Sigma).
  • PI propidium iodide
  • apoptosis analysis cells were treated as above and labeled with Annexin V-FITC followed by propidium iodide exclusion using the Annexin V-FITC Apoptosis Detection kit I (BD PharMingen, San Diego, CA) according to the manufacturer's protocols. Flow cytometry (as described in the previous Example) was used to assay both cell cycle and apoptosis studies.
  • Apoptosis analysis was carried out by flow cytometry and the percentages of cells in quadrants UL (upper left), UR (upper right), LL (lower left) and LR (lower right) were determined by CellQuest Software (Becton Dickinson).
  • AnnV Annexin V-FlTC and PI: Propidium iodide.
  • Example 26 CROIl: A naked fully human IgGl for use in melanoma therapy exploiting the mechanism of antibody-dependent cellular cytotoxicity (ADCC)
  • isotype switching from an IgG2 to an IgGl might enable the mAb to kill human melanoma cells through ADCC effector functions.
  • Human PBMC were isolated from whole blood using a Ficoll-Plaque. Briefly, in a 50 mL tube, 15 mL of PBS was added to 20 mL of whole blood which was underlayed with 10 mL Ficoll-Plaque and the tube was centrifuged at 2000 RPM. Mononuclear cells were collected from the interface and washed
  • EuTDA highly fluorescent and stable chelate
  • CROl 1 IgGl mAb to CG56972/GPNMB can kill CG56972/GPNMB expressing melanoma cells in vitro and potentially human melanoma in vivo through ADCC effector functions.
  • CROl 1 IgGl mAb can also be useful in combination with immune effector cytokines that could provide some clinical benefit in metastatic melanoma such as high dose IL-2, interferon- gamma or TNF-alpha.
  • CROl 1 can also be used to treat melanoma in combination with vaccine immunotherapy, immunomodulators such as MDX-010, radiation therapy and/or chemotherapy.
  • Example 27 Treatment of Astrocytoma, Glioblastoma, Medulloblastoma and Other tumors of the CNS
  • CG56972 is a highly drug-refractory neoplasm representing significant unmet medical needs.
  • GPNMB human gene
  • CG56972 is a type I transmembrane protein potentially involved in vesicular trafficking with a very restricted expression pattern in human brain.
  • CROl 1-vcMMAE was biochemically characterized and tested for therapeutic activity against cell lines derived from human brain tumors of astrocytoma, glioblastoma, medulloblastoma or neuroectodermal origin.
  • Transcript expression analysis demonstrated highly elevated CG56972 mRNA in brain tumors derived from astrocytoma, glioblastomas, medulloblastoma and tumors of neuroectodermal origin with restricted low expression in normal brain.
  • CROl 1 bound by FACS analysis surface CG56972 on brain cancer cell lines.
  • CROl 1 mAbs western blotted the predicted 100 and 120 kDa gene products.
  • Clonogenic assays demonstrated that CROl 1-vcMMAE mAbs inhibited the growth of brain cancer cell lines.
  • RNA samples were derived from normal human tissues obtained commercially (Clontech, Palo Alto, CA; Invitrogen, Carlsbad, CA) or cell lines grown according to specifications. RNAs were harvested and PCR was performed as previously described (Shimkets RA et. al. Nat Biotechnol., 1999. 17-8: 798-803) using TaqMan ⁇ reagents (PE Applied Biosystems, Foster City, CA). RNAs were normalized utilizing human ⁇ -actin and glyceraldehyde-3- phosphate dehydrogenase (GAPDH) TaqMan ⁇ probes according to the manufacturer's instructions.
  • GPDH glyceraldehyde-3- phosphate dehydrogenase
  • CT threshold cycle
  • CuraChipTM Tissues were lysed in Trizol. Biotin-labeled cDNA was made by using 15 mg of total RNA with poly(T) primers. Gene expression was evaluated by hybridization to the proprietary CuraChip microarray (CuraGen, New Haven, CT) of 11,000 oligonucleotide probes. Slides were hybridized for 15 h at 30 0 C with constant rotation, washed for 30 min at room temperature (RT), incubated in streptavidin solution (4°C,
  • oligonucleotide sequence used to detect CG56972 is 5'- TGATCAGTAAGGATTTCACCTCTGTTTGTA (SEQ ID NO: 341).
  • the oligonucleotide sequence used to detect GAPDH is 5'-ACCTTGTCATGTACCATCAATAAAGTACCC (SEQ ID NO: 342), corresponding to bp 1243-1272 of the GAPDH transcript (accession no. NM_002046).
  • Flow Cytometry Quantitative analysis of CG56972 expression on the surface of cell lines was determined by flow cytometry (FACS). Approximately 1 x 10 6 cells were harvested, washed and incubated with a saturating amount (10 ⁇ g/mL). of either CROIl or isotype-matched control antibody in staining buffer containing PBS (pH 7.4), 4% FBS and 0.1% NaN3 for 30 min on ice, followed by washing and staining with R-Phycoerythrin (PE)-conjugated goat-anti-human antibody (Jackson ImmunoResearch Laboratories, Inc, West Grove, PA) at 1 :100 for 30 min on ice.
  • FACS flow cytometry
  • SW-1783 cells were harvested and lysed on ice for 30 min in lysis buffer containing 1 % NP-40, 0.15 M NaCl, 0.02 M Tris-HCl, 10% glycerol, 0.01 M EDTA and complete protease inhibitor mixture (Roche Molecular Biochemicals, Indianapolis, IN). Supernatants were collected and the protein concentration was determined with the BCA Protein Assay Kit (Pierce, Rockford, IL). For immunoblot analysis, 40 ul of total cell lysate from one well of confluent cells harvested from a 6 well Falcon tissue culture dish were boiled in Laemmli sample buffer, centrifuged and resolved under reducing condition on 4-20% Tris-glycine gels (Invitrogen).
  • Clonogenic assays The growth-inhibitory activity of CRO 11 -vcMMAE was determined by clonogenic assay. Cells were plated in 96-well plates and allowed to recover overnight. CROl 1-vcMMAE or isotype-matched monoclonal antibody at various concentrations was added to sub-confluent cell cultures and incubated for 4 days at 37°C. The cells were then transferred into 6-well plates and allowed to form colonies. Colonies were stained with Giemsa stain (Sigma) and counted. The surviving cell fractions were calculated based upon the ratio of the treated sample and the untreated control. The results were expressed as a percentage of control using GraphPad Prism Version 4 software. The IC50 was defined as the concentration resulting in a 50% reduction of colony formation compared to untreated control cultures.
  • Results 1. CG56972 transcript expression in human astrocytoma, glioblastoma, medulloblastoma and tumors of neuroectodermal origin.
  • CG56972 was also expressed at high levels in 4/5 glioma human biopsies and 1/4 medulloblastoma human biopsies.
  • CG56972 was found to be highly expressed in 5/9 brain cancers of astrocytoma or glioblastoma origin as well as 4/9 oligodendrogliomas.
  • Our analysis of these tumor expression profiles showed that CG56972 message was detected to a much lesser degree in normal brain tissues.
  • These data are also consistent with our immunohistochemical data that demonstrated the lack of CROl 1 staining in normal human brain including neurons and glial cells. Taken together, these data demonstrate that the CG56972 transcript is expressed at highly elevated quantities in brain cancer and oligodendroglioma cell lines and specimens isolated from human tumors.
  • the CG56972 protein is predicted to be a type I transmembrane glycoprotein.
  • the highly elevated expression of CG56972 transcripts and the potential cell surface localization of this protein in human cancer samples encouraged us to generate monoclonal antibodies (mAbs) as a potential cancer therapeutic. Therefore, we cloned the human CG56972 extracellular domain (ECD; aa 23-480). Sequencing of the cloned cDNA revealed the presence of an in-frame 36-nt insertion, likely due to alternative splicing at the exon 6/7 boundary, which added an additional 12-aa (ATTLKS YDSNTP) (SEQ ID NO: 343) after residue 339 of the published GPNMB protein sequence.
  • CG56972 protein expression in our panel of brain cancer cell lines total cell lysates were harvest, resolved by SDS-PAGE, transferred to membrane filters and subjected to immunoblot analysis with a CG56972 polyclonal antibody.
  • the CG56972 polyclonal antibody detected two protein species that are differential glycosylation products of CG56972 of approximately 100 and 120 kDa from various brain cancer cell lines that have been shown to express CG56972 transcripts ( Figure 13A).
  • CG56972 protein was highly expressed in XP-498, SNB-78 and H79-MG and SF-539 cells.
  • CG56972 possesses a very restricted human tissue expression pattern.
  • CROl 1 did not inhibit the growth of CG56972-expressing cancer cell lines when used directly (data not shown). Since CG56972 is a cell surface molecule on brain cancers and melanoma, and since CROl 1 was internalized following incubation with CG56972-expressing cells, we evaluated whether CROI l would inhibit the growth of cancer cells when combined with a protein synthesis inhibitor (saporin)-conjugated secondaiy antibody. Our results indicated that CROl 1 could specifically inhibit the growth of
  • CROl 1-vcMMAE possessed IC50s of approximately 215, 450, 1250, and 1050 ng/mL on XF-498, SNB-78, U-118-MG and SF-539 cells ( Figure 16 and Table 54).
  • IC50s correlated with cell surface density as measured by FACS analysis.
  • conjugated control human IgG2 antibody- vcMMAE failed to inhibit the growth of any of the cell lines examined at concentrations up to 3 ⁇ g/mL (Table 54) with IC50s exceeding 1.5 or 4.5 ⁇ g/mL (Table 54).
  • Table 54 Summary of RTQ PCR, FACS and in vitro growth inhibition of human cancer cell lines with CRO 11 -mAbs
  • CT values were determined by RTQ PCR as described in Materials and Methods.
  • Geometric Mean ratios were determined by flow cytometric analysis.
  • Cytotoxicity (ADC) or cell killing was determined by clonogenic assay as described.
  • b IC 50 value is the mean and SD of a representative clonogenic assay with each experiment performed in triplicate wells. ND: Not done.

Abstract

The present invention provides fully human monoclonal antibodies that specifically bind to GPNMB, and uses thereof. Nucleotide sequences encoding, and amino acid sequences comprising, heavy and light chain immunoglobulin molecules, particularly sequences corresponding to contiguous heavy and light chain sequences spanning the framework regions and/or complementarity determining regions (CDRs) are provided. The present invention also provides immunoconjugates comprising anti-GPNMB antibodies and methods of using such immunoconjugates. The present invention further provides bispecific antibodies comprising an anti-GPNMB antibody component and an anti-CD3 component, and methods of using such bispecific antibodies.

Description

ANTIBODIES DIRECTED TO GPNMB AND USES THEREOF
FIELD OF THE INVENTION
The present invention relates to antibodies with specificity to GPNMB, and uses of such antibodies. In particular, the present invention provides fully human monoclonal antibodies that specifically bind to GPNMB, and uses thereof. Nucleotide sequences encoding, and amino acid sequences comprising, heavy and light chain immunoglobulin molecules, particularly sequences corresponding to contiguous heavy and light chain sequences spanning the framework regions and/or complementarity determining regions (CDRs) are provided. The present invention also provides immunoconjugates comprising anti-GPNMB antibodies and methods of using such immunoconjugates. The present invention further provides bi-specific antibodies comprising an anti-GPNMB antibody component and an anti-CD3 component, and methods of using such bispecific antibodies.
BACKGROUND OF THE INVENTION
GPNMB A putative transmembrane glycoprotein called "ninV (Ace. No. X76534 EMBL) ,
' referred to herein as GPNMB, was identified and described by Weterman et al., (Int J Cancer 60:73-81, 1995) as differentially expressed in low-metastatic human melanoma cancer cell lines and xenografts, compared to a more aggressive melanoma cell line. GPNMB shares 33% identity with the precursor of pMel 17 melanocyte-specific protein (Kwon et al , 1991 , PNAS 88:9228-9232). GPNMB is 71 % homologous to a dendiitic cell- associated transmembrane protein, DC-HIL (Shikano et al, 2001 Biol. Chem. 276:8125- 8134). GPNMB is also known as the hematopoietic growth factor inducible neurokinin- 1 protein HGFIN (Bandari et al, Reg. Peptides 111 :169-178) and the bone-related gene osteoactivin (Owen et al. Crit Rev Eukaryot Gene Expr 2003, 13(2-4):205-220) It was also reported that nmb could reduce the metastatic potential of a highly metastatic mnb-negative melanoma cell line (Weterman, 1995). GPNMB was considered a candidate glioblastoma tumor marker after public database mining and expression profiling (Loging et al, 2000, Genome Research 10: 1393-1402). This gene was found overexpressed in lung tumors (US Patent Publication No. US20030064947), as well as breast, rectal and colon cancers (US Patent Publication No. US2003100720). NCBI SAGE data also shows overexpression of this gene in stomach and pancreatic carcinoma. The mouse ortholog has been shown to be highly upregulated in a neural stem cell line NSC, derived from the TSC2 knockout model for Tuberous Sclerosis Complex Syndrome
(International Publication No. WO 2003/080856).
Antibodies
Antibodies, also known as immunoglobulins, are typically tetrameric glycosylated proteins composed of two light (L) chains (about 25 kDa) and two heavy (H) chains (about 50-70 kDa). The amino-terminal portion of each chain includes a variable domain of about 100 to 110 or more amino acids primarily responsible for antigen recognition. The carboxy- terminal portion of the L and H chain has one and three or four constant domains, respectively that are primarily responsible for effector function. There are two types of human L chains, classified as kappa and lambda. H chains are classified as mu, delta, gamma, alpha, or epsilon based upon the constant domain amino acid sequence, defining the antibody's isotype as IgM, IgD, IgG, IgA, and IgE, respectively. Isotypes may be further divided into subclasses e.g. IgGi, IgG2, IgG3, IgG4.
Immunoglobulins can be produced naturally in vivo by B lymphocytes. Each clone of B cells produces antibody with an antigen receptor having a unique prospective antigen binding structure. The repertoire of antigen receptors, approximately 107 possibilities, exists in vivo prior to antigen stimulation. This diversity is produced by somatic recombination, i.e., the joining of different antibody gene segments. Immunoglobulin H chain, kappa L chain and lambda L chain are encoded by three separate genetic loci and each locus has multiple copies of at least 3 types of gene segments encoding variable (V), constant (C) and joining (J) regions, the heavy chain gene also includes a diversity (D) region. The selection of specific V, C and J regions (and D for the heavy chain) from amongst the various gene segments available (45 heavy chain V; 35 kappa V; 23 heavy chain D; 6 heavy chain J; 5 kappa J) generates approximately 10 possible specificities of germline sequences exhibited in B cells. The joining of V, C and J regions can result in the loss or addition of residues at the junctions. The L and H chain V region of human antibodies consists of relatively conserved framework regions (FR) that form a scaffold for three hypervariable regions also known as complementary determining regions (CDR). From the amino terminus of either the heavy or light chain, the V domain is made up of FR and CDR regions in the following order: FRl -CDRl -FR2-CDR2-FR3. Joining of the V domain with a D (heavy chain only) and J domain adds CDR3-FR4. The CDRs are generally responsible for antigen binding. The specificity of monoclonal antibodies have made them attractive agents for targeting cancer in vivo with the hopes of eradicating disease while sparing normal tissue. The approach, which initially utilized mouse monoclonal antibodies has encountered limitations to potential effectiveness such as immunogenicity; inefficient effector functions and short half-life in vivo. Technologies were developed for: chimeric antibodies which sought to utilize the antigen binding variable domains of mouse monoclonal antibodies combined with the constant regions of human antibodies (Boulianne, et al 1984 Nature 312:643-646; Morrison et al, 1984 PNAS USA 81:6851-6855); humanized antibodies which grafted antigen binding complementary determining regions (CDRs) from mouse antibodies to human immunoglobulin (Jones, et al, 1986 Nature 321: 522-525; Riechmann, et al, 1988 Nature 332:323-327; Verhoeyen, et al, 1988 Science 239:1534-1536; Vauglum, et al, 1998 Nature Biotechnol. 16:535-539); and phage display libraries of single chain scFvs or Fab fragments of antibodies (de Haard, et al, 1999 J Biol. Chem. 274: 18218-18230; Knappik, et al, 2000 J. MoI. Biol. 296:57-86; Sheets, et al, 1998 PNAS USA 95:6157-6162; Vaughan, et al, 1994 Nature Biotechnol 14:309-314, 1996; Griffiths et al EMBO J. 13:3245-3260). Additionally, transgenic animals having human immunoglobulin genes and nonfunctional endogenous genes have been developed for immunization and production of fully human monoclonal antibodies (Fislrwild, et al, 1996 Nature Biotechnol 14:845-851; Mendez, et al, 1997 Nature Genet. 15:146-156; Nicholson, et al, 1999 J. Immunol 163, 6898-6906). Single Chain Antibodies: Single chain Fv antibodies (scFvs) were first described in the late 19801S (Bird et al, Science 242:423-426 (1988); Huston et al, Proc. Natl. Acad. Sci. USA 85:5879-5883 (1988)). A polypeptide linker, typically ranging in length from 5 to 27 amino acid residues, is used to join the C-terminus of the variable light chain domain (VL) to the N-terminus of the variable heavy chain domain (VH). Alternatively, the linker joins the C-terminus of the VH to the N-terminus of the VL- Both foπnats (VL-VH and VH- VL) have been used successfully in the literature. The most common linker used in the literature is the (Gly4Ser)3 15 amino acid linker, however there are several other linkers that have been utilized, including a 25 amino acid linker called 205C (Pantoliano et al, Biochemistry 30:10117-10125 (1991)). Single chain antibodies are currently in the clinic; one of the most advanced is h5Gl .1 or Pexelizumab. This scFv is specific for human C5 complement and is being used in clinical trials for cardiac patients undergoing cardiopulmonary bypass surgery (Shernan et al, Ann. Thorac Surg. 77:942-949 (2004)). Bispecific Antibodies (bi-Abs): An area of mAb research where considerable progress has been made is in the development of bispecific antibodies (biAbs). There are distinct advantages to developing therapeutic antibody molecules with dual specificity. For example, biAbs can serve as mediators to target immune effector cells such as CTLs to unwanted cells (Baeuerle et al., Curr. Opin. MoI. Ther. 5:413-419 (2003)). In another example, chemically linked bispecific antibodies directed against Fc gamma receptors CD 16, CD64, and CD89, were significantly more effective in vitro than conventional IgG antibodies (Peipp and Valerius, Biochem. Soc. Trans. 30:507-511 (2002)). One of the challenges in developing biAbs as viable therapeutics has been producing large enough quantities of a stable moiety for clinical applications. Another challenge has been in determining the right combination of validated targets and the underlying biology that would lead to a therapeutic product. For recent reviews on the difficulties experienced with biAbs, see (Kontermann, Acta Pharmacol Sin 26:1-9 (2005); Peipp and Valerius, Soc. Trans. 30:507-511 (2002)). Bispecific Single Chain Antibodies (bi-scFv): A notable type of biAb that can be made is a bi-specific single chain antibody or bi-scFv. For a review on the generation of bi- scFv's see (Kipriyanov and Le Gall, Curr Opin Drug Discov Devel 7:233-242 (2004)). Bi- scFvs are typically comprised of 4 variable domains, 2 heavy (VH) and 2 light (VL), which are derived from 2 different antibodies. The 4 domains are linked together with 3 short linkers, ranging in length from 5-27 amino acids. The biological activity of this type of antibody depends on several features concerning the construction of the molecule. For example, both the linker sequences between the antibody V domains and the order of the 4 antibody V domains themselves (for the 2 antibodies) can vary, as well as the expression system that is used; all of which can greatly affect the solubility and biological activity of the various resulting products (Kipriyanov et al, J. MoI. Biol. 330:99-111 (2003); Le Gall et al, Protein Eng. Des. SeI. 17:357-366 (2004); Pavlinkova et al, Clin Cancer Res. 5:2613-1619 (1999)).
Cytotoxic T lymphocytes: Under normal circumstances, T cells are activated when the CD3/T cell receptor (CD3/TCR) complex binds to a relevant MHC molecule associated with a specific Ag peptide. Engagement of CD3/TCR with MHC results in intracellular signals necessary to trigger an immune response against a pathogen or tumor. Similar signals that cause T cell activation can also be achieved by antibodies that can bind certain structures of the CD3/TCR complex. In the literature, it has been shown that biAbs recognizing both the TCR/CD3 complex and tumor associated antigen (TAA) can trigger the activation program in CTLs in the presence of target cells (Chapoval et al, J. Immunol 155:1296-1303 (1995)).
Recombinant technologies are being utilized to enable further improvements upon antibody molecules with the goal of enhancing in vivo efficacy. Such technologies provide, for example, for optimizing molecular size, affinity, pharmacokinetics, toxicity, specificity, valency, effector functions, direct and indirect arming, combination therapy, and various prodrug approaches.
It would be desirable to have an antibody suitable for in vivo targeting of GPNMB expressing pathologies and to enable therapeutic efficacy.
SUMMARY OF THE INVENTION
The current invention provides human monoclonal antibodies that specifically bind GPNMB as well as variants, derivatives and antigen binding fragments of such antibodies. The invention provides preferred somatic recombinations of human antibody gene segments to provide specificity for GPNMB and genetically engineered anti -GPNMB antibody variants and derivatives that originate from these gene segments. In addition, the current invention provides multiple affinity matured human antibodies with binding specificity for GPNMB.
In one embodiment, the present invention provides an antibody, or binding fragment thereof, that binds to GPNMB, wherein said antibody, or binding fragment thereof, neutralizes a GPNMB-induced activity, and wherein said antibody, or binding fragment thereof, cross-reacts with a fully human anti-GPNMB antibody selected from the group consisting of Mab 1.2.1, Mab 1.10.1, and Mab2.22.1 or an antibody in the same antigen- binding bin as fully human anti-GPNMB antibody Mabl .2.1 , Mabl .10.1 , or Mab2.22.1. In another embodiment, the present invention provides an antibody, or binding fragment thereof, that binds to GPNMB, wherein said antibody, or binding fragment thereof, neutralizes a GPNMB-induced activity, and wherein said antibody, or binding fragment thereof, cross-reacts with a fully human anti-GPNMB antibody selected from the group consisting of Mab2.3.1 and Mabl.15.1 or an antibody in the same antigen-binding bin as fully human anti-GPNMB antibody Mab2.3.1 or Mabl.15.1.
In yet another embodiment, the present invention provides an antibody, or binding fragment thereof, that binds to GPNMB, wherein said antibody, or binding fragment thereof, neutralizes a GPNMB-induced activity, and wherein said antibody, or binding fragment thereof, cross-reacts with fully human anti-GPNMB antibody Mab2.10.1 or an antibody in the same antigen-binding bin as fully human anti-GPNMB antibody Mab2.10.1.
In one embodiment, the present invention provides naked IgGl anti-GPNMB antibodies that have cytotoxic effect to cells overexpressing GPNMB. In a specific embodiment, the present invention provides methods of treating or preventing diseases associated with overexpression of GPNMB comprising administering to a subject in need thereof a composition comprising a naked IgGl anti-GPNMB antibody and an immunomodulator (such as, but not limited to, interferons and cytokines).
In another embodiment, the present invention provides immunoconjugates that comprise an anti-GPNMB antibody or a fragment thereof, and a cytotoxic agent. In a specific embodiment, the cytotoxic agent is auristatin E (dolastatin-10) or a derivative thereof. Methods of using such immunoconjugated are also provided.
In one embodiment, the present invention provides bispecific antibodies comprising an anti-GPNMB component and an anti-CD3 antibody component, which enable the cytotoxic killing of target tumor cells by T cells. In another embodiment, the present invention provides single chain Fv antibody conjugated to a cytotoxic agent. In a specific embodiment, the cytotoxic agent is auristatin E (dolastatin-10) or a derivative thereof. Methods of using such bispecific antibodies and conjugated single chain Fv antibodies are also provided. Amino acid sequences for anti-GPNMB human monoclonal antibodies of the invention and nucleic acid sequences encoding them are provided.
Compositions comprising human anti-GPNMB antibodies, including therapeutic compositions comprising same, and methods of use are provided. Particularly, therapeutic immunoconjugates comprising anti-GPNMB antibodies and a cytotoxic or cytostatic agent for treating GPNMB expressing cancers and other GPNMB related disorders are provided. Dosage regimens are also provided.
Additional aspects of the disclosure will be set forth in part in the description which follows, and in part will be obvious from the description, or may be learned by practicing the invention.
BRIEF DESCRIPTION OF THE FIGURES
Figure 1: Tumor growth inhibition and complete regression of SK-MEL-2 xenografts in athymic mice after treatment with 2.50 to 20 mg/kg i.v. every 4 days for 4 treatments. The responses of tumor-bearing animals to reference drugs such as vinblastine (1.7 mg/kg i.v. q4d X4) and paclitaxel (24 mg/kg i.v. q2d X4) are also shown. Control groups are treated with either phosphate-buffered saline (PBS) or physiological saline.
Figure 2: Indirect immunotoxin killing of UACC-62 melanoma cells by anti- GPNMB antibodies
Figure 3: Inhibition of colony formation of UACC-62 cells incubated with Auristatin E (AE) conjugated anti-GPNMB antibodies.
Figure 4: Tumor growth inhibition and complete regression of SK-MEL-2 xenografts in athymic mice after treatment with CROl 1-vcMMAE 5.0 mg/kg i.v. every 4 days for 4 treatments. The lack of responses of tumor-bearing animals to unconjugated CROl 1 or to free monomethylauristatin E demonstrate that the intact immunoconjugate is essential for anti-tumor effects.
Figure 5: Tumor size reduction and complete regression of SK-MEL-2 xenografts in athymic mice after treatment with 1.25 to 20 mg/kg i.v. every 4 days for 4 treatments. The responses of tumor-bearing animals to reference drugs such as Vinblastine (1.7 mg/kg i.v. q4d X4) and paclitaxel (24 mg/kg i.v. q2d X4) are also shown. Control groups are treated with either phosphate-buffered saline (PBS) or physiological saline.
Figure 6: The serum concentration-time profile of the antibody of CROl 1-vcMMAE after intravenous administration of 1 and 10 mg/kg in athymic mice. Detection was achieved with a sandwich ELISA assay, which employed the CROl 1 antigen (CG56972,
GPNMB) and a horseradish peroxidase-conjugated anti-human globulin. Results shown are the serum concentrations expressed as μg/mL (left x-axis) and micromolarmolar concentration (right X-axis).
Figure 7: Aggregate responses, expressed as percent cures, were recorded for test animals treated with 5 different, graduated dosing intervals (i.e., 0, 1, 4, 8, and 16 days between treatments). The slope of the line is not significantly different from 0 (p< 0.2904).
Figure 8: The proportions of complete regressors as a function of dosing interval and stratified by cumulative dose. For each group, n= 6 mice/group. Athymic mice bearing established SK-MEL-2 tumor implants (day 14, 80 mg) were treated i.v. with CROIl- vcMMAE and the incidence of complete regressions is recorded.
Figure 9: Effects of ectopic expression of GPNMB or sensitivity to CROIl- vcMMAE. HEK293 cells are transfected with empty vector (vector) or GPNMB-containing plasmid (GPNMB) as described in Materials and Methods. A. Cell lysates are prepared from the transfected HEK293 cells and the expression of GPNMB (upper panel) or actin (lower panel) is determined by immunoblotting. Lane 1: Empty vector transfectants. Lane 2: GPNMB transfectants. B. Flow cytometry analysis of GPNMB expression on empty vector or GPNMB transfected cells. C. CROl 1-vcMMAE in vitro growth inhibition of transfected cells. Cells are treated with various concentrations of CRO 11 -vcMMAE (diamonds: vector or circles: GPNMB) or IgG2-vcMMAE (triangles: vector or squares: GPNMB) for 96 hours. After a clonogenic assay, the surviving fraction is normalized to the untreated control and expressed as a percentage of the control using GraphPad Prism graphing software. Each treatment is performed in triplicate. A representative graph from two independent experiments is shown.
Figure 10: Effect of GPNMB siRNA on endogenous GPNMB expression and sensitivity to CRO 11 -vcMMAE. SK-Mel-2 cells are transfected with 50 nJVI of control siRNA or siRNA tai'geting GPNMB. A. Cell Iy sates are prepared from the transfected SK- Mel-2 cells 2 and 4 days post-transfection and the expression of GPNMB (upper panel) or actin (lower panel) is determined by immunoblotting. Lane 1 : Mock (oligofectamine) transfection. Lane 2: Control siRNA transfection. Lane 3: GPNMB siRNA transfection. B. Flow cytometry analysis of GPNMB expression 2 and 4 days after transfection. SK-Mel-2 cells are transfected with mock, control siRNA or GPNMB siRNA as indicted in the Materials and Methods. C. CROl 1 -vcMMAE in vitro growth inhibition of mock (diamonds), control siRNA (circles) or GPNMB siRNA (triangles) transfected SK-Mel-2 cells is determined by a clonogenic assay as described in Materials and Methods. The surviving fraction is normalized to the untreated control and expressed as a percentage of control using GraphPad Prism graphing software. Each treatment is performed in triplicate. A representative experiment from two independent studies is shown. Figure 11: FACS analysis of SK-MEL-2 with isotype control, hybridoma IgG2
(B2), recombinant IgG2 (B19) and recombinant IgGl (B16) to CG56972/GPNMB relative to IgG2 (B2, B19) or IgGl (Control, B16) controls.
Figure 12: (A) PBMC and mAb (IgGl) mediated ADCC of SK-MEL-2 cells. ADCC effector functions are measured as described above at 2, 5 and 10 μg/200 μl using targeteffector ratios of 10, 30, 60 and 100 as indicated. (B) PBMC and mAb (IgG2) do not cause ADCC to SK-MEL-2 cells. ADCC effector functions are measured as described above at 0, 2, 5 and 10 μg/200 μl using target: effector ratios of 10, 30, 60 and 100 as indicated. Figure 13: Expression of CG56972 in human cancer cell lines and tissues. RTQ PCR analysis of (A) human brain cancer cell lines or (B) human brain cancer glioma and medulloblastoma biopsies. (C) Microarray analysis of CG56972 expression in human brain cancer and oligodendroglioma tissues. Tissues or cell lines are harvested, mRNA prepared and RTQ PCR or CuraChip analysis performed as described in Materials and Methods.
Figure 14: FACS analysis of cell surface binding of CROl 1 mAb to CG56972. SK- MEL-2, XF-498, U-118-MG, SNB-78, SF-539 and SF-268 cells are labeled with a saturating concentration (10 μg/mL) of CROl 1 mAb or control IgG2. Bound mAb is detected by flow cytometry with PE-conjugated goat-anti-human secondary antibody as described in Materials and Methods. GM: Geometric mean. The SF-268 cell line is CG56972 transcript negative and used as a negative control.
Figure 15: Immunoblot analysis of CG5672 expression in human brain cancer cell lines. Cell lysates are resolved on Tris-glycine gels and transferred to membranes. Immunoblot analysis is carried out with a polyclonal antibody to CG56972 followed by enhanced chemiluminescence detection as described in Materials and Methods. Arrowheads indicate the relative mobility of the plOO and 120 CG56972 species. The SF-268 cell line is CG56972 transcript negative and used as a negative control.
Figure 16: CROl 1-vcMMAE in vitro growth inhibition of astocytoma/glioblastoma cell growth. XF-498, SNB-78, U-118-MG, SF-539, LOXIMVI and SF-268 cells are incubated with the indicated concentration of CROl 1-vcMMAE. Cells are also incubated with control PKl 6.3 mAb (data shown in Table I) as described in the Materials and Methods. Cell growth was determined by clonogenic assay. The surviving colonies are counted and plotted using GraphPad Prism graphing software. The experiment is performed in triplicate wells and repeated twice. vA representative experiment is shown. IC50s for cell killing is presented in ng/mL concentrations. The LOXIMVI and SF-268 cell lines are CG56972 transcript negative and used as negative controls.
Figure 17: Development of CROI l Engineered Antibodies. Four antibody variable (V) domains (shown in C for the bi-scFv) are derived from the light and heavy chain variable domains (VL and VH) making up the antigen binding sites of CROl 1 and anti-CD3 whole IgGs. The middle linker joining the 2 individual scFv components together (shown in dashed line) may play a key role in determining the resulting activity of each of the scFv components, including the effective cytolytic activity provided by the cytotoxic T cells engaged by the anti-CD3 scFv component of the bi-scFv. Figure 18: A. ELISA results for CROl 1 scFv (squares) and CROl 1 x anti-CD3 (L4- L2-L4 linker set) bi-scFv (diamonds). Both engineered CROl 1 antibodies bound to the GPNMB target. B. Western blotting of 2 of the CROI l engineered antibody products (arrows). Clone 16 corresponded to the CHOKl line expressing CROl 1 scFv (monomer), while clone 17 corresponded to the CHOKl line expressing CROl 1 x anti-CD3 (L4-L2-L4 linker set) bi-scFv (dimer). Clones 16 and 17 are used to produce the engineered antibody products.
Figure 19: Flow cytometry analysis of binding of CROl 1 scFv and CROl 1 x anti- CD3 (L4-L2-L4 linker set) bi-scFv products to native GPNMB protein expressed on the cell surface of target cells. Human T cells are used as a source of CD3, while SK-Mel-5 cells are used as a source of GPNMB.
Figure 20: Cytotoxicity analysis showed that purified CROl 1 x anti-CD3 (L4-L2-L4 linker set) bi-scFv, but not CROl 1 scFv, causes killing of GPNMB positive SK-Mel-5 tumor cells by T lymphocytes. Figure 21: The chemical structure of Maleimidocoaproyl-Valine-Citrullin-
Monomethyl-Auristatin E (vcMMAE).
Figure 22: Disulfides on CROl 1 antibody are gently reduced in the presence of TCEP to generate ~4 thiols per Ab. vcMMAE is then added to antibody solution. Nucleophilic attack of thiolates on maleimide-groups results in a stable thioester linkage. The resulting conjugate is purified from the mixture.
Figure 23: Reaction of vcMMAE with NAcCys at pH 7.0 and pH 9.0 in the presence or absence of TCEP. IA: VCMMAE converts fully into NAcCys-adduct following a incubation in phosphate pH 7 buffer. B-E: Appearance of a side product in a course of incubation of vcMMAE in borate buffer. F-I: Appearance of side products in borate pH 9 and in the presence of TCEP.
Figure 24: LCMS identification of the side product with retention time of 9.2 min not capable of reaction with cystein and therefore, not capable of conjugation to CROl 1.
Figure 25: Kinetics of the formation of NAcCys-vcMMAE and of the side product (succinimidyl-vcMMAE) following incubation in borate pH 9.0 buffer in the presence or absence of TCEP. DETAILED DESCRIPTION OF THE INVENTION
As used herein, the term "antibody" refers to an immunoglobulin or a fragment or a derivative thereof, and encompasses any polypeptide comprising an antigen-binding site, regardless whether it is produced in vitro or in vivo. The term includes, but is not limited to, polyclonal, monoclonal, monospecific, polyspecific, non-specific, humanized, single-chain, chimeric, synthetic, recombinant, hybrid, mutated, engineered, and grafted antibodies. Unless otherwise modified by the term "intact," as in "intact antibodies," for the purposes of this disclosure, the term "antibody" also includes antibody fragments such as Fab, F(ab')2, Fv, scFv, bi-scFv, bi-Ab, Fd, dAb, and other antibody fragments that retain antigen-binding function, i.e., the ability to bind GPNMB specifically. Typically, such fragments would comprise an antigen-binding domain.
As used herein, the terms "antigen-binding domain," "antigen-binding fragment," and "binding fragment" refer to a part of an antibody molecule that comprises amino acids responsible for the specific binding between the antibody and the antigen. In instances, where an antigen is large, the antigen-binding domain may only bind to a part of the antigen. A portion of the antigen molecule that is responsible for specific interactions with the antigen-binding domain is referred to as "epitope" or "antigenic determinant."
An antigen-binding domain typically comprises an antibody light chain variable region (VL) and an antibody heavy chain variable region (VH), however, it does not necessarily have to comprise both. For example, a so-called Fd antibody fragment consists only of a VH domain, but still retains some antigen-binding function of the intact antibody.
As used herein, the term "repertoire" refers to a genetically diverse collection of nucleotides derived wholly or partially from sequences that encode expressed immunoglobulins .-The sequences are generated by in vivo rearrangement of, e.g., V, D, and J segments for H chains and, e.g., V and J segment for L chains. Alternatively, the sequences may be generated from a cell line by in vitro stimulation, in response to which the rearrangement occurs. Alternatively, part or all of the sequences may be obtained by combining, e.g. , unrearranged V segments with D and J segments, by nucleotide synthesis, randomised mutagenesis, and other methods, e.g., as disclosed in U.S. Pat. No.5,565,332. As used herein, the terms "specific interaction" and "specific binding" refer to two molecules forming a complex that is relatively stable under physiologic conditions. Specific binding is characterized by a high affinity and a low to moderate capacity as distinguished from nonspecific binding which usually has a low affinity with a moderate to high capacity. Typically, binding is considered specific when the affinity constant KA is higher than 106 M" 1 , or more preferably higher than 108 M"1. If necessary, non-specific binding can be reduced without substantially affecting specific binding by varying the binding conditions. The appropriate binding conditions such as concentration of antibodies, ionic strength of the solution, temperature, time allowed for binding, concentration of a blocking agent (e.g., serum albumin, milk casein), etc., may be optimized by a skilled artisan using routine techniques.
As used herein, the term "substantially as set out" refers that the relevant CDR, VH, or VL domain of the invention will be either identical to or have only insubstantial differences in the specified regions (e.g., a CDR), the sequence of which is set out. Insubstantial differences include minor amino acid changes, such as substitutions of 1 or 2 out of any 5 amino acids in the sequence of a specified region.
As used herein, the term "CROl 1" refers to a fully human monoclonal antibody that specifically binds to GPNMB. In some embodiments, CROl 1 refers to those antibodies that are identified in Tables 2A-2D of the present application. In some embodiments, CROl 1 refers to Mab 1.15.1 as described in the instant invention.
The terras "GPNMB" and "CG56972" are used interchangeably herein. As used herein, the terms "GPNMB" or "CG56972" refer to a transmembrane glycoprotein that has an amino acid sequence as set forth in SEQ ID NO: 289, an analog, derivative or a fragment thereof, or a fusion protein comprising GPNMB, an analog, derivative or a fragment thereof. In certain embodiments, the term "GPNMB" refers to the mature, processed form of GPNMB. In other embodiments, the term "GPNMB" refers to the extracellular domain of GPNMB. As used herein, the term "GPNMB activity" refers to one or more activities associated with GPNMB. To "modulate" GPNMB activity is to alter the baseline results observed with, and that can be attributed to GPNMB. To "neutralize" GPNMB is to cancel one or more effects, e.g. activity observed with, and that can be attributed to GPNMB.
As used herein, the term "isolated" refers to a molecule that is substantially free of its natural environment. For instance, an isolated protein is substantially free of cellular material or other proteins from the cell or tissue source from which it is derived. The term "isolated" also refers to preparations where the isolated protein is sufficiently pure to be administered as a pharmaceutical composition, or at least 70-80% (w/w) pure, more preferably, at least 80-90% (w/w) pure, even more preferably, 90-95% pure; and, most preferably, at least 95%, 96%, 97%, 98%, 99%, or 100% (w/w) pure.
As used herein, the term "inhibit" or "inhibition of refers to reducing by a measurable amount, or to prevent entirely. As used herein, the term "Cytotoxic effect" in reference to the effect of an agent on a cell, means killing of the cell. "Cytostatic effect'" refers to an inhibition of cell proliferation. A "cytotoxic agent" refers an agent that has a cytotoxic or cytostatic effect on a cell, thereby depleting or inhibiting the growth of, respectively, cells within a cell population.
As used herein, the teπns "prevent," "preventing," and "prevention" refer to the inhibition of the development or onset of a disorder associated with aberrant expression and/or activity of GPNMB (e.g., cancer) or the prevention of the recurrence, onset, or development of one or more symptoms of a disorder associated with aberrant expression and/or activity of GPNMB (e.g., cancer) in a subject resulting from the administration of a therapy or the administration of a combination of therapies. As used herein, the term "effective amount" refers to a dosage or amount that is sufficient to reduce the activity of GPNMB to result in amelioration of symptoms in a patient or to achieve a desired biological outcome.
As used herein, the term "prophylactically effective amount" refers to the amount of a therapy which is sufficient to result in the prevention of the development, recurrence, or onset of a disorder associated with aberrant expression and/or activity of GPNMB (e.g., cancer) or one or more symptoms thereof, or to enhance or improve the prophylactic effect(s) of another therapy.
As used herein, a "protocol" includes dosing schedules and dosing regimens. The protocols herein are methods of use and include prophylactic and therapeutic protocols. As used herein, the terms "subject" and "patient" are used interchangeably. As used herein, the terms "subject" and "subjects" refer to an animal, preferably a mammal including a non-primate (e.g., a cow, pig, horse, cat, dog, rat, and mouse) and a primate (e.g., a monkey, such as a cynomolgous monkey, chimpanzee, and a human), and more preferably a human. As used herein, the terms "therapeutic agent" and "therapeutic agents" refer to an agent that can be used in the prevention, treatment, management, or amelioration of a disorder associated with aberrant expression and/or activity of GPNMB (e.g., cancer) or one or more symptoms thereof. In certain embodiments, the term "therapeutic agent" refers to an antibody that immunospecifically binds to GPNMB. In certain other embodiments, the term "therapeutic agent" refers an agent other than an antibody that immunospecifieally binds to GPNMB.
As used herein, the terms "therapies" and "therapy" can refer to any protocol(s), method(s), and/or agent(s) that can be used in the prevention, treatment, management, or amelioration of a disorder associated with aberrant expression and/or activity of GPNMB (e.g., cancer) or one or more symptoms thereof. In certain embodiments, the terms "therapies" and "therapy" refer to anti-cancer therapy, biological therapy, supportive therapy, and/or other therapies useful in treatment, management, prevention, or amelioration of cancer or one or more symptoms thereof known to one of skill in the art such as medical personnel.
As used herein, the terms "treat," "treatment," and "treating" refer to the eradication, removal, modification, or control of primary, regional, or metastatic cancer tissue, or the reduction or amelioration of the progression, severity, and/or duration of a disorder associated with aberrant expression and/or activity of GPNMB or amelioration of one or more symptoms thereof resulting from the administration of one or more therapies. In certain embodiments, such terms in the context of cancer refer to a reduction in the growth of cancerous cells, a decrease in number of cancerous cells and/or a reduction in the growth, formation and/or volume of a tumor. In other embodiments, such terms refer to the minimizing or delay of the spread of cancer resulting from the administration of one or more therapies to a subject with such a disease. Treatment can include, for example, a decrease in the severity of a sypmtopm, the number of symptoms, or frequency of relapse. Unless otherwise defined, scientific and technical terms used in connection with the invention described herein shall have the meanings that are commonly understood by those of ordinary skill in the art. Further, unless otherwise required by context, singular terms shall include pluralities and plural terms shall include the singular. Generally, nomenclatures utilized in connection with, and techniques of, cell and tissue culture, molecular biology, and protein and oligo- or polynucleotide chemistry and hybridization described herein are those well known and commonly used in the art. Standard techniques are used for recombinant DNA, oligonucleotide synthesis, and tissue culture and transformation (e.g., electroporation, lipofection). Enzymatic reactions and purification techniques are performed according to manufacturer's specifications or as commonly accomplished in the art or as described herein. The foregoing techniques and procedures are generally performed according to conventional methods well known in the art and as described in various general and more specific references that are cited and discussed throughout the present specification. (See e.g., Sambrook et a Molecular Cloning: A Laboratory Manual, 2d ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, K Y. 1989). The nomenclatures utilized in connection with, and the laboratory procedures and techniques of, analytical chemistry, synthetic organic chemistry, and medicinal and pharmaceutical chemistry described herein are those well known and commonly used in the art. Standard techniques are used for chemical syntheses, chemical analyses, pharmaceutical preparation, formulation, and delivery, and treatment of patients. The current invention provides germline human antibody heavy chain V, D, J combinations and light chain V, J combinations including nucleotide and amino acid sequence of the VH and VL domain FR and CDR regions with specificity for GPNMB.
Upon exposure to antigen, those B cells with antigen binding specificity based on germline sequences are activated, proliferate, and differentiate to produce immunoglobulins of different isotypes as well as undergo somatic mutation and/or affinity maturation to produce immunoglobulins of higher affinity for the antigen. The current invention provides the nucleotide and amino acid sequence of such affinity matured V domain FR and CDR regions having specificity to GPNMB.
Fab type antibody fragments containing the antigen binding portion of the antibody molecule may consist of the L chain covalently linked by a disulfide bond to a portion of the H chain which has the V domain and first constant domain. Single chain Fv antibody fragment (scFv) has the H variable domain linked to the L variable domain by a polypeptide linker. The invention provides antibody fragments such as Fab and scFv molecules having sequences derived from germline or affinity matured V domains of antibodies binding specifically to GPNMB.
A bispecific or bifunctional antibody is an artificial hybrid antibody having two different heavy/light chain pairs and two different binding sites. Bispecific antibodies can be produced by a variety of methods including fusion of hybridomas or linking of Fab* fragments (see, e.g., Songsivilai & Lachmann, 1990 Clin. Exp. Immunol. 79: 315-321; Kostelny et ah, 1992 J. Immunol. 148:1547-1553). Bispecific antibodies do not exist in the form of fragments having a single binding site (e.g., Fab, Fab\ and Fv).
It will be appreciated that such bifunctional or bispecific antibodies are contemplated and encompassed by the invention. A bispecific single chain antibody with specificity to GPNMB and to the CD3 antigen on cytotoxic T lymphocytes can be used to direct these T cells to tumor cells expressing GPNMB and cause apoptosis and eradication of the tumor. Bispecific scFv constructs for this purpose are described herein. The scFv components specific for GPNMB can be derived from anti-GPNMB antibodies described herein. In some embodiments, the anti-GPNMB antibody components disclosed herein can be used to generate a biologically active scFv directed against GPNMB. The anti-CD3 scFv component of the therapeutic bispecific scFv was derived from a sequence deposited in Genbank (accession number CAE85148). Alternative antibodies known to target CD3 or other T cell antigens may similarly be effective in treating malignancies when coupled with anti-GPNMB, whether on a single-chain backbone or a full IgG.
GPNMB binding human antibodies may include H or L constant domains including L kappa or lambda constant regions, or any isotype H constant domain. In one embodiment of the invention, a human antibody with binding specificity to GPNMB contains germline sequences such as the heavy chain V regions: VH 1-2 (SEQ ID NO: 308), VH2-5 (SEQ ID NO: 360), VH3-11 (SEQ ID NO: 361), VH3-21 (SEQ ID NO: 362), VH3-30 (SEQ ID NO:363), VH3-33 (SEQ ID NO: 364), VH4-31 (SEQ ID NO: 365), VH4-59 (SEQ ID NO:366) or VH5-51 (SEQ ID NO:367); the heavy chain D region: Dl-20 (amino acid sequences translated by SEQ ID NO: 375), Dl -26 (amino acid sequences translated by SEQ ID NO:376), D3-10 (amino acid sequences translated by SEQ ID NO:377), D3-16 (amino acid sequences translated by SEQ ID NO:378), D3-22 (amino acid sequences translated by SEQ ID NO: 379), D3-9 (amino acid sequences translated by SEQ ID NO:380), D4-17 (amino acid sequences translated by SEQ ID NO: 381), D5-24 (amino acid sequences translated by SEQ ID NO: 382), D6-13 (amino acid sequences translated by SEQ ID NO:383), or D6-19 (amino acid sequences translated by SEQ ID NO: 384); the heavy chain J region: JH3b (SEQ ID NO: 385), JH4b (SEQ ID NO:386), JH5b (SEQ ID NO: 387) or JH6b (SEQ ID NO: 388); the light chain V kappa regions A2 (SEQ ID NO:373), A3 (SEQ ID NO: 371), A20 (SEQ ID NO: 370), A27 (SEQ ID NO: 369), A30 (SEQ ID NO:374), L2 (SEQ ID NO:372) or Ol (SEQ ID NO: 368); and the J region JKl (SEQ ID NO:389), JK2 (SEQ ID NO: 390), JK3 (SEQ ID NO: 391), JK4 (SEQ ID NO: 392) or JK5 (SEQ ID NO: 393). (generally, see Rabat Sequences of Proteins of Immunological Interest, National Institutes of Health, Bethesda, Md. 1987 and 1991; also see Chothia & Lesk 1987 J. MoI. Biol. 196:901-917; Chothia et al. 1989 Nature 342:878-883). In a particular embodiment of the invention human antibodies with binding specificity to GPNMB are combined germline regions as shown in Table 1.
TABLE 1: Human anti-GPNMB antibody germline region combinations.
Figure imgf000019_0001
In an embodiment of the invention, the isolated antibody has a heavy chain variable region polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NOs:2, 20, 38, 56, 74, 92, 110, 128, 146, 164, 182, 200, 218, 236, 253, 256, 260, 265, 270, 274, 277, 281 and 285. Such amino acid sequences can be encoded by nucleotide sequences selected from the group consisting of SEQ ID NOs: 1, 19, 37, 55, 73, 91, 109, 127, 145, 163, 181, 199, 217 and 235. In another embodiment, the invention provides an isolated antibody that specifically binds to GPNMB and has a light chain variable region polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 11, 29, 47, 65, 83, 101, 119, 137, 155, 173, 191, 209, 227 and 245. Such amino acid sequences can be encoded by nucleotide sequences selected from the group consisting of SEQ ID NOs: 10, 28, 46, 64, 82, 100, 118, 136, 154, 172, 190, 208, 226 and 244. In yet another embodiment, the invention provides an isolated antibody that specifically binds to GPNMB and has a heavy chain polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 2, 20, 38, 56, 74, 92, 110, 128, 146, 164, 182, 200, 218, 236, 253, 256, 260, 265, 270, 274, 277, 281 and 285 and has a light chain polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 11, 29, 47, 65, 83, 101, 119, 137, 155, 173, 191, 209, 227 and 245. In yet another embodiment of the invention, anti-GPNMB antibodies comprise at least one CDR of any of the H or L CDR polypeptide sequences SEQ ID NOs: 4, 6, 8, 13, 15, 17, 22, 24, 26, 31, 33, 35, 40, 42, 44, 49, 51, 53, 58, 60, 62, 67, 69, 71, 76, 78, 80, 85, 87, 89, 94, 96, 98, 103, 105, 107, 112, 114, 116, 121, 123, 125, 130, 132, 134, 139, 141, 143, 148, 150, 152, 157, 159, 161, 166, 168, 170, 175, 177, 179, 184, 186, 188, 193, 195, 197, 202, 204, 206, 211, 213, 215, 220, 222, 224, 229, 231, 233, 238, 240, 242, 247, 249, 251, 254, 257, 261, 266, 271, 278, 282, 286, 255, 258, 262, 267, 272, 275, 279, 283, 287, 259, 263, 264, 268, 269, 273, 276, 280, 284 and 288.
In particular embodiments, human anti-GPNMB antibodies are Mabl.10.2, Mabl.15.1, Mabl.2.2, Mabl.7.1, Mab2.10.2, Mab2.15.1, Mab2.16.1, Mab2.17.1, Mab2.21.2, Mab2.22.1, Mab2.24.1, Mab2.3.1, Mab2.7.1, and Mab2.8.1. These antibodies have amino acid sequences and nucleic acid sequences encoding them identified in this application as shown in Tables 2A-2D. TABLE 2A Antibod Nucleotide DNA) and Amino Acid (AA Sequences
Figure imgf000020_0001
TABLE 2B: Antibod Nucleotide (DNA and Amino Acid AA) Se uences
Figure imgf000021_0001
TABLE 2D: Antibody Nucleotide (DNA) and Amino Acid (AA) Sequences
Figure imgf000022_0001
VH4-31 derived anti-GPNMB Antibodies: In a particular embodiment, GPNMB-binding human antibodies of the invention comprise germline V heavy chain region VH4-31 or are derived therefrom and have an amino acid sequence of the formula:
X1SGPGLVKPSQX2LSLTCTVS GGSIS SX3X4YX5WX6 WIRX7HPGKGLEWIG YIYYSGX8TYX9NPSLKS RVXIOISVDTSKNQFSLXΠLSSVTAADTA VYYCAR Where: Xl is E or Q;
X2 is T or N;
X3 is A, F or G;
X4 is N or G;
X5 is Y or F; X6 is T or S;
X7 is Q or H;
X8 is S or N;
X9 is C, S or Y;
XlO is I or T; XI l is K or T;
(SEQ ID NO:253 ).
In specific embodiments SEQ ID NO:253 is combined with D3-22 or Dl -20. Furthermore the combination of SEQ ID NO:253 with D3-22 or Dl -20 is combined with JH6b or JH4b and in specific embodiments, after affinity maturation these GPNMB-binding human antibodies, for example Mab 1.15.1, Mabl.7.1 and Mab2.22.1, have amino acid sequences SEQ ID NOs:20, 56 and 164 and can be encoded by nucleotide sequences SEQ
ID NO: 19, 55 and 163. Furthermore, in particular embodiments H chain CDRl sequences are the germline VH4-31 CDR or affinity matured sequences thereof, of the formula: CDRh GGSIS SX3X4YX5WX6
Where: X3 is A, F or G; X4 is N or G;
X5 is Y or F; X6 is T or S; (SEQ ID NO:254 ). In specific embodiments an anti-GPNMB antibody of the invention comprise a
CDRl sequence selected from the following: SEQ ID NO:22, 58, 166.
In particular embodiments H chain CDR2 sequences are the germline VH4-31 CDR or affinity matured sequences thereof of the formula:
CDR2: Y[YYSGX8TYX9NPSLIVS Where: X8 is S or N;
X9 is C, S or Y; (SEQ ID NO:255).
In specific embodiments an anti-GPNMB antibody of the invention comprise a CDR2 sequence selected from the following: SEQ ID NO: 24, 60, and 168.
In particular embodiments, the H chain CDR3 sequence is a D3-22, JH6b combination having SEQ ID NO: 170. Alternatively, in particular embodiments the H chain
CDR3 sequence is a Dl-20, JH4b combination having SEQ ID NO:26 or 62.
VHl -2 derived anti-GPNMB Antibodies: In a particular embodiment, GPNMB-binding human antibodies of the invention comprise geπnline V heavy chain region VHl -2 or are derived therefrom and include an amino acid sequence of the formula:
QLVQSGAEVKBCPGASVKVSCKAS GYTFT GX1YMH WVRQX2PGQGLEWMG WINPNSGGTX3YX4QKFQX5 RVTMTRDTSISTX6YMELSRLRSDDTAVYYCAR Where: Xl is Y or F;
X2 is A or T; X3 is N or Y; X4 is A or V; X5 is D or G; X6 is A or V;
(SEQ ID NO: 256).
In specific embodiments SEQ ID NO:256 is combined with D3-10 or D6-19. Furthermore the combination ov SEQ ID NO:256 with D3-10 or D6-19 is combined with JH4b or JH5b and in specific embodiments, after affinity maturation these GPNMB-binding human antibodies, for example Mab2.3.1 and Mab 2.17.1 have amino acid sequences: SEQ ID NO:128 and 200 and can be encoded by nucleotide sequences SEQ ID NO:127 and 199. Furthermore, in particular embodiments H chain CDRl sequences are the germline VH 1-2 CDR or affinity matured sequences thereof, of the formula:
CDRl: GYTFTGXIYMH
Where: X l is Y or F, (SEQ ID NO:257 )
In specific embodiments an anti-GPNMB antibody of the invention comprise a CDRl sequence selected from SEQ ID NO: 130 and 202.
In particular embodiments H chain CDR2 sequences are the germline VH 1-2 CDR or affinity matured sequences thereof of the formula:
CDR2: WINPNSGGTX3YX4QKFQX5 Where: X3 is N or Y; X4 is A or V; X5 is D or G (SEQ ID NO:258).
In specific embodiments an anti-GPNMB antibody of the invention comprise a CDR2 sequence selected from SEQ ID NO: 132 and 204.
In particular embodiments H chain CDR3 sequences are germline D3-10, JH4b combinations or affinity matured sequences thereof, having the amino acid sequence of the formula:
CDR3: XiX2X3GSGSX4X5
Where: Xl is Y or D; X2 is Y or F; X3 is Y or F;
X4 is Y or L; X5 is Y or L (SEQ ID NO:259). In specific embodiments an anti-GPNMB antibody of the invention comprise a
CDR3 sequence selected from SEQ ID NO: 134 and 206.
VH2-5 derived anti-GPNMB Antibodies:
In a particular embodiment, GPNMB-binding human antibodies of the invention comprise germline V heavy chain region VH2-5 or are derived therefrom and include an amino acid sequence of the formula:
ITLKESGPTLVX1 PTQTLTLTCTFS GFSLS X2X3GX4GVG WIRQPPGICALX5WLX6 LIYWNDDICX7YSPSLX8S RLTITKDTSICNQVVLX9X10 TNMDPVDTATYYCAH Where: Xl is K or T;
X2 is T or A; X3 is S or G;
X4 is M or V;
X5 is D or E;
X6 is A or T;
X7 is R or H; XS is K or R;
X9 is T or R; X lO is M or I; (SEQ ID NO:260 ).
In specific embodiments SEQ ID NO:260 is combined with D3-9 or D3-16 and furthermore is combined with JH4b. In specific embodiments, after affinity maturation these GPNMB-binding human antibodies, for example, Mab 2.8.1 and Mab 1.2.2 have amino acid sequences SEQ ID NO: 38 and 236 and can be encoded by nucleotide sequences SEQ ID NO: 37 and 235.
Furthermore, in particular embodiments H chain CDRl sequences are the germline VH2-5 CDR or affinity matured sequences thereof, of the formula:
CDRl: GFSLS X2X3GX4GVG Where: X2 is T or A;
X3 is S or G;
X4 is M or V; (SEQ 1D NO:261 ).
In specific embodiments an anti-GPNMB antibody of the invention comprise a CDRl sequence selected from SEQ ID NO: 40 and 238.
In particular embodiments H chain CDR2 sequences are the germline VH2-5 CDR2 or affinity matured sequences thereof of the formula:
CDR2: LIYWNDDKX7YSPSLX8S Where: X7 is R or H;
X8 is K or R;
(SEQ ID NO:262 ).
In specific embodiments an anti-GPNMB antibody of the invention comprise a
CDR2 sequence selected from SEQ ID NO:42 and 240.
In particular embodiments H chain CDR3 sequences are germline D3-9, JH4b combinations or affinity matured sequences thereof and include an amino acid sequence of the formula:
CDR3: X1YDILTGX2X3 Where: Xl is Y or H;
X2 is Y or F; and X3 is Y or N (SEQ ID NO-.263 ).
In a specific embodiments an anti-GPNMB antibody of the invention comprises a CDR3 amino acid sequence SEQ ID NO:242.
In yet another particular embodiment H chain CDR3 sequences are germline D3-16, JH4b combinations or affinity matured sequences thereof and include an amino acid sequence of the formula:
CDR3:YDYX,WGS Where: X 1 is V or D
(SEQ ID NO:264).
In a specific embodiment an anti-GPNMB antibody of the invention comprises a CDR3 amino acid sequence SEQ ID NO: 44. VH3-33 derived anti-GPNMB Antibodies:
In a particular embodiment, GPNMB-binding human antibodies of the invention comprise germline V heavy chain region VH3-33 or are derived therefrom and have an amino acid sequence of the formula:
QVQLX 1 X2SGGGVVQPGRSLRLSCAAS GFTFX3X4YGX5H WVRQAPGKGLEWVA VI WX6DGX7NKYYADSVKG RFTISRDNSKNTLYLQMNSLRAEDXgAVYYCAXc Where: X 1 is V or E;
X2 is E or Q; X3 is S or N;
X4 is S or N;
X5 is M or I;
X6 is Y or F;
X7 is S or R; XS is T or A;
X9 is R or K
(SEQ ID NO:265).
In specific embodiments SEQ ID NO:265 is combined with D3-10 or D4-17 and furthermore with JH4b. In specific embodiments, after affinity maturation these GPNMB- binding human antibodies, for example Mab 2.7.1 and Mab2.15.1 have amino acid sequences: SEQ ID NO: 92 and 218 and can be encoded by nucleotide sequences SEQ ID NO:91 and 217.
Furthermore, in particular embodiments H chain CDRl sequences are the geπnline VH3-33 CDR or affinity matured sequences thereof, of the formula:
CDRl: GFTFX3X4YGX5H Where: X3 is S or N;
X4 is S or N;
X5 is M or I; (SEQ ID NO-.266).
In specific embodiments an anti-GPNMB antibody of the invention comprise a CDRl amino acid sequence selected from SEQ ID NO:94 and 220.
In particular embodiments H chain CDR2 sequences are the germline VH3-33 CDR2 or affinity matured sequences thereof of the formula:
CDR2: VIWX6DGX7NKYYADSVKG Where: X6 is Y or F; X7 is S or R; (SEQ ID NO:267). In specific embodiments an anti-GPNMB antibody of the invention comprise a CDR2 sequence selected from SEQ ID NO:96 and 222.
In particular embodiments H chain CDR3 sequences are D3-10, JH4b combinations or affinity matured sequences thereof and include an amino acid sequence of the formula: CDR3: YYYGSGX,
Where: Xl is S or L
(SEQ ID NO:268).
A specific embodiment is anti-GPNMB antibody 2.7.1 having a CDR3 amino acid sequence SEQ ID NO:224.
In an alternative embodiment H chain CDR3 sequences are D4-17, JH4b combinations or affinity matured sequences thereof and include an amino acid sequence of the formula:
CDR3: DYGDX1 Where: Xl is Y or S
(SEQ ID NO:269). A specific embodiment is anti-GPNMB antibody 2.15.1 having a CDR3 amino acid sequence SEQ ID NO: 98.
VH3-11 derived anti-GPNMB Antibodies:
In a particular embodiment, GPNMB-binding human antibodies of the invention comprise germline V heavy chain region VH3-11 or are derived therefrom and have an amino acid sequence of the formula:
QVQLVESGGGLVKPGGSLRLSCAAS GFTFS X1YX2MX3 WIRQAPGKGLEWVS YISX4SGSX5X6X7YADSVICG RFTX8SRDNAKNSLYLQMNSLRAEDTAVYYCAR
Where: Xl is D or S; X2 is S or Y; X3 is S or T; X4 is S or I; X5 is T or I;
X6 is T or I; X7 is Y or H; X8 is I or M; (SEQ ID NO:270).
In specific embodiments SEQ ID NO:270 is combined with D6-13 and furthermore with JH3b. In specific embodiments, after affinity maturation these GPNMB-binding human antibodies, for example Mab 2.16.1 have amino acid sequence SEQ ID NO: 110 and can be encoded by nucleotide sequence SEQ ID NO: 109. Furthermore, in particular embodiments H chain CDRl sequences are the germline
VEB -11 CDRl or affinity matured sequences thereof, of the formula:
CDRh GFTFS X1YX2MX3 Where: Xl is D or S; X2 is S or Y; X3 is S or T; (SEQ ID NO:271 ). In specific embodiments an anti-GPNMB antibody of the invention comprise a
CDRl amino acid sequence SEQ ID NO:112.
In particular embodiments H chain CDR2 sequences are the germline VH3-11 CDR2 or affinity matured sequences thereof of the formula:
CDR2: YISX4SGSX3X6X7YADSVKG Where: X4 is S or I;
X5 is T or I;
X6 is T or I;
X7 is Y or H;
(SEQ ID NO:272). In specific embodiments an anti-GPNMB antibody of the invention comprises a
CDR2 sequence SEQ ID NO:114.
In particular embodiments H chain CDR3 sequences are D6-13, JH3b combinations or affinity matured sequences thereof and include an amino acid sequence of the formula:
CDR3: X1X2AAAG- - AFDI Where: XI is G or D;
X2 is I or G; (SEQ ID NO:273).
A specific embodiment is anti-GPNMB antibody 2.16.1 having a CDR3 amino acid sequence SEQ ID NO: 116.
VH3-21 derived anti-GPNMB Antibodies:
In a particular embodiment, GPNMB-binding human antibodies of the invention comprise germline V heavy chain region VH3-21 or are derived therefrom and have an amino acid sequence of the formula:
X1VQLX2X3SGGGLVKPGGSLRX4 SCAAS GFTFS SYSMN WVRQAPGKGLEWVS X5ISS SSSYIYYADSVKG RFTISRDNAKNSLYLQMNSLRAEDTAVYYCAR Where: Xl is E or Q;
X2 is V or E; X3 is E or Q;
X4 is F or L;
X5 is S or F;
(SEQ ID N 0:274). In specific embodiments SEQ ID NO:274 is combined with D 1-26 and furthermore with JH4b. In specific embodiments, after affinity maturation these GPNMB-binding human antibodies, for example Mab 2.21.1 have amino acid sequence SEQ ID NO: 146 and can be encoded by nucleotide sequence SEQ ID NO: 145.
Furthermore, in particular embodiments H chain CDRl sequences are the germline VH3-21 CDRl, SEQ ID NO: 148 or affinity matured sequences thereof.
In particular embodiments H chain CDR2 sequences are the germline VH3-21 CDR2 or affinity matured sequences thereof of the formula:
CDR2: X5ISS SSSYIYYADSVKG Where: X5 is S or F;
(SEQ ID NO:275). In specific embodiments an anti-GPNMB antibody of the invention comprises a
CDR2 amino acid sequence SEQ ID NO: 150.
In particular embodiments H chain CDR3 sequences are D 1-26, JH4b combinations or affinity matured sequences thereof and include an amino acid sequence of the formula:
CDR3: X1X2VGAT-FDY Where: X l is G or D;
X2 is I or W;
(SEQ ID NO:276). A specific embodiment is anti-GPNMB antibody 2.21.1 having a CDR3 amino acid sequence SEQ ID NO: 152.
VH3-30 derived anti-GPNMB Antibodies:
In a particular embodiment, GPNMB-binding human antibodies of the invention comprise germline V heavy chain region VH3-30 or are derived therefrom and include an amino acid sequence of the formula:
QLVESGGGVVQPGRSLRLSCAAS GFX1FS SYGMH WVRQAPGKGLEWVA VISYDGX2NKYYADSVKG RFTISRDNSKNTLYLQMNSLRAEDTAVYYCAK Where: XI is T or A; X2 is S or N;
(SEQ ID NO:277).
In specific embodiments SEQ ID NO:277 is combined with D3-10 and furthermore with JH6b. In specific embodiments, after affinity maturation these GPNMB-binding human antibodies, for example Mab 2.10.2 have amino acid sequence SEQ ID NO:74 and can be encoded by nucleotide sequence SEQ ID NO:73.
Furthermore, in particular embodiments H chain CDRl sequences are the germline VH3-30 CDRl, or affinity matured sequences thereof having an amino acid sequence of the formula: GFXiFS SYGMH
Where: Xl is T or A;
(SEQ ID NO:278). In specific embodiments an anti-GPNMB antibody of the invention comprise a
CDRl sequence SEQ ID NO:76. In particular embodiments H chain CDR2 sequences are the germline VΗ3-30
CDR2 or affinity matured sequences thereof of the formula: CDR2: VISYDGX2NKyYADSVKG Where: X2 is S or N;
(SEQ ID NO:279). In specific embodiments an anti-GPNMB antibody of the invention comprises a CDR2 amino acid sequence SEQ ID NO:78.
In particular embodiments H chain CDR3 sequences are D3-10, JH 6b combinations or affinity matured sequences thereof and include an amino acid sequence of the formula:
Figure imgf000030_0001
Where: Xl is I or D; X2 is T or L;
X3 is M or V; X4 is V or 1; X5 is I or R; X6 is I or G; (SEQ 1D NO:280).
A specific embodiment is anti-GPNMB antibody 2.10.2 having a CDR3 amino acid sequence SEQ ID NO:80.
VH4-59 derived anti-GPNMB Antibodies:
In a particular embodiment, GPNMB-binding human antibodies of the invention comprise germline V heavy chain region VH4-59 or are derived therefrom and include an amino acid sequence of the formula:
QVQLQESGPGLVKPSETLSLTCTVS GX1SIS X2YYWS WIRQPPGKGLEWIG YXjYYSGSTNYNPSLKS RVTISVDTSKNQFSLKLSSVTAADTAVYYCAR
Where: Xl is G or D; X2 is S or N; X3 is I or F; (SEQ ID NO:2S1).
In specific embodiments SEQ ID NO:281 is combined with D6-19 and furthermore with JH4b. In specific embodiments, after affinity maturation these GPNMB-binding human antibodies, for example Mab 1.10.2 have amino acid sequence SEQ ID NO:2 and can be encoded by nucleotide sequence SEQ ID NO:1. Furthermore, in particular embodiments H chain CDRl sequences are the germline
VH4-59 CDRl, or affinity matured sequences thereof having an amino acid sequence of the formula:
GX1SIS X2YYWS Where: Xl is G or D; X2 is S or M;
(SEQ ID NO:282). In specific embodiments an anti-GPNMB antibody of the invention comprise a CDRl sequence SEQ ID NO:4. In particular embodiments H chain CDR2 sequences are the germline VH4-59 CDR2 or affinity matured sequences thereof of the formula:
CDR2: YXjYYSGSTNYNPSLKS Where: X3 is I or F; (SEQ ID NO:283).
In specific embodiments an anti-GPNMB antibody of the invention comprises a
CDR2 amino acid sequence SEQ ID NO:6.
In particular embodiments H chain CDR3 sequences are D6-19, JH4b combinations or affinity matured sequences thereof and include an amino acid sequence of the formula: CDR3: X1X2GW-DY
Where: Xl is S or D; X2 is S or R; (SEQ ID NO:284). A specific embodiment is anti-GPNMB antibody 1.10.2 having a CDR3 amino acid sequence SEQ ID NO:8. VH5-51 derived anti-GPNMB Antibodies:
In a particular embodiment, GPNMB-binding human antibodies of the invention comprise germline V heavy chain region VH5-51 or are derived therefrom and include an amino acid sequence of the formula:
QLVQSGAEVKKPGESLKISCX1GS GYX2FT X3YWIG WVRQMPGKGLEWMG X4IYPX5DSDTRYSPSFQG QVTISADKSISTAYLQWSSLKASDTAX6YYCAR Where: Xl is K or Q; X2 is S or I;
X3 is S or N; X4 is I or V; X5 is G or D; X6 is M or I; (SEQ ID NO:285).
In specific embodiments SEQ ID NO:285 is combined with D5-24 and furthermore with JH4b. In specific embodiments, after affinity maturation these GPNMB-binding human antibodies, for example Mab 2.24.1 have amino acid sequence SEQ ID NO: 182 and can be encoded by nucleotide sequence SEQ ID NO: 181.
Furthermore, in particular embodiments H chain CDRl sequences are the germline VH5-51 CDRl, or affinity matured sequences thereof having an amino acid sequence of the formula:
GYX2FT X3YWIG Where: X2 is S or I;
X3 is S or N; (SEQ ID NO:286). In specific embodiments an anti-GPNMB antibody of the invention comprise a CDRl sequence SEQ ID NO: 184.
In particular embodiments H chain CDR2 sequences are the germline VH5-51 CDR2 or affinity matured sequences thereof of the formula: CDR2: X4IYPX5DSDTRYSPSFQG
Where: X4 is I or V; X5 is G or D; (SEQ ID NO:287). In specific embodiments an anti-GPNMB antibody of the invention comprises a CDR2 amino acid sequence SEQ ID NO: 186.
In particular embodiments H chain CDR3 sequences are D5-24, JH4b combinations or affinity matured sequences thereof and include an amino acid sequence of the formula:
CDR3: X,WLQX2--FDY Where: Xl is R or K; X2 is L or H;
(SEQ 1D NO:288).
A specific embodiment is anti-GPNMB antibody 2.24.1 having a CDR3 amino acid sequence SEQ ID NO: 188. The antibodies of the invention bind an epitope of GPNMB (SEQ ID NO:289), preferably within the mature sequence of GPNMB and more preferably within the extracellular domain (ECD) of GPNMB.
Antibodies of the invention bind GPNMB with an affinity of 10'6 to 10~n . Preferably with an affinity of 10~7 or greater and even more preferably 10" or greater. In a preferred embodiment, antibodies described herein bind to GPNMB with very high affinities (Kd), for example a human antibody that is capable of binding GPNMB with a Kd less than, but not limited to, 1(T7, 10"8, 1(T9, 10"10, 1(T11, 1(T12, ICT13 or 10"14 M, or any range or value therein. Affinity and/or avidity measurements can be measured by KinExA® and/or BIACORE®, as described herein. In particular embodiments antibodies of the invention bind to GPNMB with Kds ranging from 50 to 150 pM.
Epitope mapping and secondary and tertiary structure analyses can be carried out to identify specific 3D structures assumed by the disclosed antibodies and their complexes with antigens (see, e.g., Epitope Mapping Protocols, ed. Morris, Humana Press, 1996). Such methods include, but are not limited to, X-ray crystallography (Biochem. Exp. Biol, 11: 7-13, 1974) and computer modeling of virtual representations of the presently disclosed antibodies (Fletterick et al. (1986) Computer Graphics and Molecular Modeling, in Current Communications in Molecular Biology, Cold Spring Harbor Laboratory, Cold Spring Harbor, N. Y).
Furthermore, the specific part of the protein immunogen recognized by antibody may be determined by assaying the antibody reactivity to parts of the protein, for example an N terminal and C terminal half. The resulting reactive fragment can then be further dissected, assaying consecutively smaller parts of the immunogen with the antibody until the minimal reactive peptide is defined. Alternatively, the binding specificity, that is the epitope, of anti-GPNMB antibodies of the invention may be determined by subjecting GPNMB immunogen to SDS-PAGE either in the absence or presence of a reduction agent and analyzed by immunoblotting. Epitope mapping may also be performed using SELDI. SELDI ProteinChip® (LumiCyte) arrays used to define sites of protein-protein interaction. GPNMB protein antigen or fragments thereof may be specifically captured by antibodies covalently immobilized onto the PROTEINCHIP array surface. The bound antigens may be detected by a laser-induced desorption process and analyzed directly to determine their mass.
The epitope recognized by anti-GPNMB antibodies described herein may be determined by exposing the PROTEINCHIP Array to a combinatorial library of random peptide 12-mer displayed on Filamentous phage (New England Biolabs). Antibody-bound phage are eluted and then amplified and taken through additional binding and amplification cycles to enrich the pool in favor of binding sequences. After three or four rounds, individual binding clones are further tested for binding by phage ELISA assays performed on antibody-coated wells and characterized by specific DNA sequencing of positive clones. Derivatives
This disclosure also provides a method for obtaining an antibody specific for GPNMB. CDRs in such antibodies are not limited to the specific sequences of H and L variable domains identified in Table 1 and may include variants of these sequences that retain the ability to specifically bind GPNMB. Such variants may be derived from the sequences listed in Table 1 by a skilled artisan using techniques well known in the art. For example, amino acid substitutions, deletions, or additions, can be made in the FRs and/or in the CDRs. While changes in the FRs are usually designed to improve stability and immunogenicity of the antibody, changes in the CDRs are typically designed to increase affinity of the antibody for its target. Variants of FRs also include naturally occurring immunoglobulin allotypes. Such affinity-increasing changes may be determined empirically by routine techniques that involve altering the CDR and testing the affinity of the antibody for its target. For example, conservative amino acid substitutions can be made within any one of the disclosed CDRs. Various alterations can be made according to the methods described in the art (Antibody Engineering, 2.sup.nd ed., Oxford University Press, ed. Borrebaeck, 1995). These include but are not limited to nucleotide sequences that are altered by the substitution of different codons that encode a functionally equivalent amino acid residue within the sequence, thus producing a "silent" change. For example, the nonpolar amino acids include alanine, leucine, isoleucine, valine, proline, phenylalanine, tryptophan, and methionine. The polar neutral amino acids include glycine, serine, threonine, cysteine, tyrosine, asparagine, and glutamine. The positively charged (basic) amino acids include arginine, lysine, and histidine. The negatively charged (acidic) amino acids include aspartic acid and glutamic acid. Substitutes for an amino acid within the sequence may be selected from other members of the class to which the amino acid belongs (see Table 3). Furthermore, any native residue in the polypeptide may also be substituted with alanine (Acta Physiol. Scand. Suppl. 643:55-67, 1998; Adv. Biophys. 35:1-24, 1998).
TABLE 3. Amino acid substitutions
Figure imgf000034_0001
Derivatives and analogs of antibodies of the invention can be produced by various techniques well known in the art, including recombinant and synthetic methods (Maniatis (1990) Molecular Cloning, A Laboratory Manual, 2.sup.nd ed., Cold Spring Harbor Laboratory, Cold Spring Harbor, N. Y., and Bodansky et al. (1995) The Practice of Peptide Synthesis, 2.sup.iιd ed., Spring Verlag, Berlin, Germany).
Preferred amino acid substitutions are those which: (1) reduce susceptibility to proteolysis, (2) reduce susceptibility to oxidation, (3) alter binding affinity for forming protein complexes, (4) alter binding affinities, and (4) confer or modify other physicochemical or functional properties of such analogs. Analogs can include various muteins of a sequence other than the naturally-occurring peptide sequence. For example, single or multiple amino acid substitutions (preferably conservative amino acid substitutions) may be made in the naturally-occurring sequence (preferably in the portion of the polypeptide outside the domain(s) forming intermolecular contacts). A conservative amino acid substitution should not substantially change the structural characteristics of the parent sequence {e.g., a replacement amino acid should not tend to break a helix that occurs in the parent sequence, or disrupt other types of secondary structure that characterizes the parent sequence). Examples of art-recognized polypeptide secondary and tertiary structures are described in the art (for example, Proteins, Structures and Molecular Principles (Creighton, Ed., W. H. Freeman and Company, New York (1984)).
In one embodiment, a method for making an H variable domain which is an amino acid sequence variant of an H variable domain of the invention comprises a step of adding, deleting, substituting, or inserting one or more amino acids in the amino acid sequence of the presently disclosed H variable domain, optionally combining the H variable domain thus provided with one or more L variable domains, and testing the H variable domain or H variable/L variable combination or combinations for specific binding to GPNMB or and, optionally, testing the ability of such antigen-binding domain to modulate GPNMB activity. The L variable domain may have an amino acid sequence that is identical or is substantially as set out according to Table 1.
An analogous method can be employed in which one or more sequence variants of a L variable domain disclosed herein are combined with one or more H variable domains.
A further aspect of the disclosure provides a method of preparing antigen-binding fragment that specifically binds with GPNMB. The method comprises: (a) providing a starting repertoire of nucleic acids encoding a H variable domain that either includes a
CDR3 to be replaced or lacks a CDR3 encoding region; (b) combining the repertoire with a donor nucleic acid encoding an amino acid sequence substantially as set out herein for a H variable CDR3 such that the donor nucleic acid is inserted into the CDR3 region in the repertoire, so as to provide a product repertoire of nucleic acids encoding a H variable domain; (c) expressing the nucleic acids of the product repertoire; (d) selecting a binding fragment specific for GPNMB; and (e) recovering the specific binding fragment or nucleic acid encoding it. Again, an analogous method may be employed in which a L variable CDR3 of the invention is combined with a repertoire of nucleic acids encoding a L variable domain, which either include a CDR3 to be replaced or lack a CDR3 encoding region. The donor nucleic acid may be selected from nucleic acids encoding an amino acid sequence substantially as set out in SEQ ID NOs: 2, 20, 38, 56, 74, 92, 110, 128, 146, 164, 182, 200, 218, 236, 253, 256, 260, 265, 270, 274, 277, 281, 285, 1 1 , 29, 47, 65, 83, 101, 1 19, 137,
155, 173, 191, 209, 227 and 245. A sequence encoding a CDR of the invention {e.g., CDR3) may be introduced into a repertoire of variable domains lacking the respective CDR {e.g., CDR3), using recombinant DNA technology, for example, using methodology described by Marks et al. {Bio/Technology (1992) 10: 779-783). In particular, consensus primers directed at or adjacent to the 5' end of the variable domain area can be used in conjunction with consensus primers to the third framework region of human H variable genes to provide a repertoire of H variable domains lacking a CDR3. The repertoire may be combined with a CDR3 of a particular antibody. Using analogous techniques, the CDR3-derived sequences may be shuffled with repertoires of H variable or L variable domains lacking a CDR3, and the shuffled complete H variable or L variable domains combined with a cognate L variable or H variable domain to make the GPNMB specific antibodies of the invention. The repertoire may then be displayed in a suitable host system such as the phage display system such as described in WO92/01047 so that suitable antigen-binding fragments can be selected. Analogous shuffling or combinatorial techniques may be used (e.g. Stemmer, Nature
(1994) 370: 389-391). In further embodiments, one may generate novel H variable or L variable regions carrying one or more sequences derived from the sequences disclosed herein using random mutagenesis of one or more selected H variable and/or L variable genes, such as error-prone PCR {Proc. Nat. Acad. ScL U.S.A. (1992) 89: 3576-3580). Another method that may be used is to direct mutagenesis to CDRs of H variable or L variable genes {Proc. Nat. Acad. ScL U.S.A. (1994) 91: 3809-3813; J. MoI. Biol. (1996) 263: 551-567). Similarly, one or more, or all three CDRs may be grafted into a repertoire of H variable or L variable domains, which are then screened for an antigen-binding fragment specific for GPNMB.
A portion of an immunoglobulin variable domain will comprise at least one of the CDRs substantially as set out herein and, optionally, intervening framework regions as set out herein. The portion may include at least about 50% of either or both of FRl and FR4, the 50% being the C-terminal 50% of FRl and the N-terminal 50% of FR4. Additional residues at the N-terminal or C-terminal end of the substantial part of the variable domain may be those not normally associated with naturally occurring variable domain regions. For example, construction of antibodies by recombinant DNA techniques may result in the introduction of N- or C-terminal residues encoded by linkers introduced to facilitate cloning or other manipulation steps. Other manipulation steps include the introduction of linkers to join variable domains to further protein sequences including immunoglobulin heavy chain constant regions, other variable domains (for example, in the production of diabodies), or proteinaceous labels as discussed in further detail below. Although the embodiments illustrated in the Examples comprise a "matching" pair of H variable and L variable domains, a skilled artisan will recognize that alternative embodiments may comprise antigen-binding fragments containing only a single CDR from either L variable or H variable domain. Either one of the single chain specific binding domains can be used to screen for complementary domains capable of forming a two- domain specific antigen-binding fragment capable of, for example, binding to GPNMB. The screening may be accomplished by phage display screening methods using the so-called hierarchical dual combinatorial approach disclosed in WO92/01047, in which an individual colony containing either an H or L chain clone is used to infect a complete library of clones encoding the other chain (L or H) and the resulting two-chain specific binding domain is selected in accordance with phage display techniques as described.
Anti-GPNMB antibodies described herein can be linked to another functional molecule, e.g., another peptide or protein (albumin, another antibody, etc.), toxin, radioisotope, cytotoxic or cytostatic agents. For example, the antibodies can be linked by chemical cross-linking or by recombinant methods. The antibodies may also be linked to one of a variety of nonproteinaceous polymers, e.g. , polyethylene glycol, polypropylene glycol, or polyoxyalkylenes, in the manner set forth in U.S. Pat. Nos. 4,640,835; 4,496,689; 4,301,144; 4,670,417; 4,791,192; or 4,179,337. The antibodies can be chemically modified by covalent conjugation to a polymer, for example, to increase their circulating half-life. Exemplary polymers and methods to attach them are also shown in U.S. Pat. Nos. 4,766,106; 4,179,337; 4,495,285, and 4,609,546.
The disclosed antibodies may also be altered to have a glycosylation pattern that differs from the native pattern. For example, one or more carbohydrate moieties can be deleted and/or one or more glycosylation sites added to the original antibody. Addition of glycosylation sites to the presently disclosed antibodies may be accomplished by altering the amino acid sequence to contain glycosylation site consensus sequences known in the art. Another means of increasing the number of carbohydrate moieties on the antibodies is by chemical or enzymatic coupling of glycosides to the amino acid residues of the antibody (WO 87/05330; CRC Crit. Rev. Biochem., 22: 259-306, 1981). Removal of any carbohydrate moieties from the antibodies may be accomplished chemically or enzymatically (Arch. Biochem. Biophys., 259: 52, 1987; Anal. Biochem., 118: 131, 1981; Meth. Enzymol, 138: 350, 1987). The antibodies may also be tagged with a detectable, or functional, label. Detectable labels include radiolabels such as 131I or 99Tc, which may also be attached to antibodies using conventional chemistry. Detectable labels also include enzyme labels such as horseradish peroxidase or alkaline phosphatase. Detectable labels further include chemical moieties such as biotin, which may be detected via binding to a specific cognate detectable moiety, e.g., labeled avidin.
The valency of the antibodies may be custom designed to affect affinity and avidity, retention time at binding sites (see e.g. Am H. Pathol, 2002 160:1597-1608; J. Med. Chem. 2002 45:2250-2259; Br. J. Cancer 2002 86:1401-1410; Biomol. Eng. 2001 18:95-108; Int J. Cancer 2002 100:367-374).
Multiple specificity (bifunctional) binding reagents may be designed based upon the GPNMB specific sequences of the invention (Biomol. Eng.2001 18:31-40). For example, a bispecific or bifunctional antibody is an artificial hybrid antibody having two different heavy/light chain pairs and two different binding sites. Bispecific antibodies can be produced by a variety of methods including fusion of hybridomas or linking of Fab' fragments (Clin. Exp. Immunol. 1990, 79: 315-321;, J. Immunol. 199,2148:1547-1553). Such bispecific antibodies can be generated comprising a specificity to GPNMB and a second specificity to a second molecule using techniques that are well known (Immunol Methods 1994,4:72-81; Wright and Harris, supra. ; Traunecker et all 992 Int. J. Cancer (Suppl.) 7:51-52). Bispecific antibodies prepared in this manner selectively kill cells expressing GPNMB. Antibodies, in which CDR sequences differ only insubstantially from those set out in SEQ ID NOs: 4, 6, 8, 13, 15, 17, 22, 24, 26, 31, 33, 35, 40, 42, 44, 49, 51, 53, 58, 60, 62, 67, 69, 71, 76, 78, 80, 85, 87, 89, 94, 96, 98, 103, 105, 107, 1 12, 1 14, 116, 121, 123, 125, 130, 132, 134, 139, 141, 143, 148, 150, 152, 157, 159, 161, 166, 168, 170, 175, 177, 179, 184, 186, 188, 193, 195, 197, 202, 204, 206, 21 1, 213, 215, 220, 222, 224, 229, 231, 233, 238, 240, 242, 247, 249 and 251. And formulas: 254, 257, 261, 266, 271, 278, 282, 286, 255, 258, 262, 267, 272, 275, 279, 283, 287, 259, 263, 264, 268, 269, 273, 276, 280, 284, 288, are encompassed within the scope of this invention. Typically, an amino acid is substituted by a related amino acid having similar charge, hydrophobic, or stereochemical characteristics. Such substitutions would be within the ordinary skills of an artisan. Unlike in CDRs, more substantial changes can be made in FRs without adversely affecting the binding properties of an antibody. Changes to FRs include, but are not limited to engineering certain framework residues that are important for antigen contact or for stabilizing the binding site, e.g., changing the class or subclass of the constant region, changing specific amino acid residues which might alter the effector function such as Fc receptor binding (U.S. Pat. Nos. 5,624,821; 5,648,260;Lund et al. (1991) J. Immiin. 147: 2657 -2662 ;Morgan et al. (1995) Immunology 86: 319-324), or changing the species from which the constant region is derived.
One of skill in the art will appreciate that the derivatives and modifications described above are not all-exhaustive, and that many other modifications would be obvious to a skilled artisan in light of the teachings of the present disclosure. Nucleic Acids, Cloning and Expression Systems
The present disclosure further provides isolated nucleic acids encoding the disclosed antibodies. The nucleic acids may comprise DNA or RNA and may be wholly or partially synthetic or recombinant. Reference to a nucleotide sequence as set out herein encompasses a DNA molecule with the specified sequence, and encompasses a RNA molecule with the specified sequence in which U is substituted for T, unless context requires otherwise. The nucleic acids provided herein comprise a coding sequence for a CDR, a H variable domain, and/or a L variable domain disclosed herein. The present disclosure also provides constructs in the foπn of plasmids, vectors, phagemids, transcription or expression cassettes which comprise at least one nucleic acid encoding a CDR, a H variable domain, and/or a L variable domain disclosed here. The disclosure further provides a host cell which comprises one or more constructs as above.
Also provided are nucleic acids encoding any CDR (CDRl, CDR2, CDR3 from either the H or L variable domain), H variable or L variable domain, as well as methods of making of the encoded products. The method comprises expressing the encoded product from the encoding nucleic acid. Expression may be achieved by culturing under appropriate conditions recombinant host cells containing the nucleic acid. Following production by expression, a H variable or L variable domain, or specific binding member may be isolated and/or purified using any suitable technique, then used as appropriate. Antigen-binding fragments, FI variable and/or L variable domains and encoding nucleic acid molecules and vectors may be isolated and/or purified from their natural environment, in substantially pure or homogeneous form, or, in the case of nucleic acid, free or substantially free of nucleic acid or genes of origin other than the sequence encoding a polypeptide with the required function. Systems for cloning and expression of a polypeptide in a variety of different host cells are well known in the art including cells suitable for producing antibodies {Gene Expression Systems, Academic Press, eds. Fernandez et al., 1999). Briefly, suitable host cells include bacteria, plant cells, mammalian cells, and yeast and baculovirus systems. Mammalian cell lines available in the art for expression of a heterologous polypeptide include Chinese hamster ovary (CHO) cells, HeLa cells, baby hamster kidney cells, NSO mouse myeloma cells, and many others. A common bacterial host is E, coli. Any protein expression system compatible with the invention may be used to produce the disclosed antibodies. Suitable expression systems also include transgenic animals {Gene Expression Systems, Academic Press, eds. Fernandez et at, 1999). Suitable vectors can be chosen or constructed, so that they contain appropriate regulatory sequences, including promoter sequences, terminator sequences, polyadenylation sequences, enhancer sequences, marker genes and other sequences as appropriate. Vectors may be plasmids or viral, e.g., phage, or phagemid, as appropriate {see Sambrook et al., Molecular Cloning: A Laboratory Manual, Lsup.nd ed., Cold Spring Harbor Laboratory? Press, 1989). Many known techniques and protocols for manipulation of nucleic acid, for example, in preparation of nucleic acid constructs, mutagenesis, sequencing, introduction of DNA into cells and gene expression, and analysis of proteins, are known in the art {Current Protocols in Molecular Biology, 2. sup. nd Edition, eels. Ausubel et al, John Wiley & Sons, 1992).
The invention also provides a host cell comprising a nucleic acid as disclosed herein. A still further aspect provides a method comprising introducing such nucleic acid into a host cell. The introduction may employ any available technique. For eukaryotic cells, suitable techniques may include calcium phosphate transfection, DEAE-Dextran, electroporation, liposome-mediated transfection and transduction using retrovirus or other virus, e.g. , vaccinia or, for insect cells, baculovirus. For bacterial cells, suitable techniques may include calcium chloride transformation, electroporation and transfection using bacteriophage. The introduction of the nucleic acid into the cells may be followed by causing or allowing expression from the nucleic acid, e.g., by culturing host cells under conditions for expression of the gene. Immunoconjugates
In another aspect, the antibodies of the invention can be used as a targeting agent for delivery of another therapeutic or a cytotoxic agent to a cell expressing GPNMB. The method includes administering an anti-GPNMB antibody coupled to a therapeutic or a cytotoxic agent or under conditions that allow binding of the antibody to GPNMB.
Anti-GPNMB antibodies are conjugated to a therapeutic agent, such as a cytotoxic compound, such that the resulting immimoconjugate exerts a cytotoxic or cytostatic effect on a GPNMB expressing cell. Particularly suitable moieties for conjugation to antibodies are chemotherapeutic agents, prodrug converting enzymes or toxins. For example, an anti- GPNMB antibody can be conjugated to a cytotoxic agent such as a chemotherapeutic agent (see infra) or a toxin (e.g. abrin, ricin A, pseudomonas exotoxin, or diphtheria toxin). Alternatively, anti-GPNMB antibody may be conjugated to a pro-drug converting enzyme. The pro-drug converting enzyme can be recombinantly fused to the antibody or derivative thereof or chemically conjugated thereto using known methods. Examplary pro-drug converting enzymes are carboxypeptidase G2, β-glucuronidase, penicillin- V-amidase, penicillin-G-amidase, β-lactamase, β-glucosidase, nitroreductase and carboxypeptidase A. Any agent that exerts a therapeutic effect on GPNMB expressing cells can be used as an agent for conjugation to an anti-GPNMB antibody of the invention. Useful classes of cytotoxic agents include, for example, antitubulin agents, auristatins, DNA minor groove binders, NDA replication inhibitiors, alkylating agents (e.g., platinum complexes such as cis-plantin, mono(platinum), bis(platinum) and tri-nuclear platinum complexes and carboplatin), anthracyclines, antiboiotics, antifolates, antimetabilites, chemotherapy sensitizers, duocarmycins, etoposides, fluorinated purimidines, ionophores, lexitropsins, nitrosoureas, platinols, pre-forming compounds, purine antimetabolites, puromcins, radiation sensitizers, steroids, taxanes, topoisomerase inhibitors, vinca alkaloids, or the like. The therapeutic agent can be a cytotoxic agent. Suitable cytotoxic agents include, for example, dolastatins (e.g. auristatin E, AFP, MMAF, MMAE), DNA minor groove binders (e.g., enediynes and lexitropsins), cuocarmycins, taxanes (e.g., paclitaxel and docetaxel), puromycins, vinca alkaloids, CC- 1065, SN-38, topotecan, morpholino- doxorubicin, rhizoxin, cyanomorpholino-doxorubicin, echinomycin, combretastatin, netropsin, epothilone A and B, estramustine, cryptophysins, cemadotin, maytansinoids, discodeπnolide, eleutherobin, and mitoxantrone.
In a specific embodiment, the cytotoxic or cytostatic agent is auristatin E (dolastatin- 10) or a derivative thereof (e.g. an ester formed between auristatin E and a keto acid). Other typical auristatin derivatives include AFP, MMAR, and MMAE. The synthesis and structure of auristatin E and its derivates are described in U.S. Patent Application
Publication No. 20030083263; PCT/US03/24209; PCT/US02/13435; and U.S. Patent Nos. 6,323,315; 6,239,104; 6,034065; 5,780,588; 5,665,860; 5,663,149; 5,635,483; 5,599,902; 5,554,725; 5,530,097; 5,521,284; 5,504,191; 5,410,024; 5,138,036; 5,076,973; 4,986,988; 4,978,744; 4,879,278; 4,816,444; and 4,486,414. In a specific embodiment anti-GPNMB antibody 1.15.1 was coupled to monomethylauristatin E via intracellular protease-sensitive valine-citruUine peptide linker (vcMMAE). Methods for making the immunoconjugate can be found in Doronina S.O. et al, 2003 Nature Biotechnology 21(7):778-794.
Techniques for conjugating therapeutic agents to proteins, and in particular, antibodies are known in the art (see, e.g. Arnon et al, 1985 in Monoclonal Antibodies and Cancer Therapy, Reisfeld et al. eds., Alan R. Liss, Inc., 1985; Hellstrom et al., 1987 in Controlled Drag Delivery, Robinson et al. eds., Marcel Dekker, Inc., 2nd ed. 1987; Thorpe 1985, in Monoclonal Antibodies '84: Biological and Clinical Applications, Pinchera et al. eds., EDITOR, 1985; Monoclonal Antibodies for Cancer Detection and Therapy, Baldwin el al. eds., Academic Press 1985; and Thorpe et al, 1982, Immunol. Rev. 62:119-58). In certain embodiments of the invention, anti-GPNMB antibodies binding to GPNMB expressing cells, are internalized and accumulate in the cell. Thereby anti-GPNMB antibody immunoconjugates accumulate in GPNMB expressing cells. Typically when the anti-GPNMB antibody immunoconjugate is internalized, the agent is preferentially active. Alternatively, anti GPNMB immunoconjugates are not internalized and the drug is effective to deplete or inhibit GPNMB expressing cells by binding to the cell membrane. The therapeutic agent can be conjugated in a manner that reduces its activity unless it is cleaved off the antibody (e.g. by hydrolysis or by a cleaving agent). In this case, the agent can be attached to the antibody or derivative thereof with a cleavable linker that is sensitive to cleavage in the intracellular environment of the target but is not substantially sensitive to the extracellular environment, such that the conjugate is cleaved from the antibody or derivative thereof when it is internalized by the GPNMB expressing cell (e.g. in the endosomal or, for example by virtue of pH sensitivity or protease sensitivity, in the lysosomal environament or in a caveolea).
A therapeutic agent of the immunoconjugate can be charged relative to the plasma membrane (e.g. polarized or net charge relative to the plasma membrane), thereby further minimizing the ability of the agent to cross the plasma membrane once internalized by a cell.
The anti-GPNMB antibody immunoconjugate can comprise a linker region between the therapeutic agent and the antibody. The linker can be cleavable under intracellular conditions, such that cleavage of the linker releases the therapeutic agent from the antibody in the intracellular environment. The linker can be, e.g. a peptidyl linker that is cleaved by an intracellular peptidase or protease enzyme, including but not limited to a lysosomal or endosomal protease. Often the peptidyl linker is at least two amino acids long or at least three amino acids long. Cleaving agnets can include cathepsins and D and plasmin, all of which are known to hydrolyze dipeptide drug derivative s resulting in the release of active drug inside target cells (see Dubowchik and Walker, 1999 Pharm. Therapuetics 83:67-123). Other linkers are described e.g. in U.S. Patent No. 6,214,345.
Linkers can be pH-sensitive can often be hydrolizable under acidic conditions such as is found in the lysosome (see e.g. U.S. Patent Nos. 5,122,368; 5,824,805; 5,622,929; Dubowchik and Walker, 1999 Pharm. Therapuetics 83:67-123; Neville et ah, 1989 BIoI. Chem. 264:14653-14661). Such linkers are relatively stable under neutral pH conditions, such as those in the blood, but are unstable at below pH 5.5 or 5.0, the pH of the lysosome. Linkers can be cleavable under reducing conditions (e.g. a disulfide linker) (see e.g., Thorpe et ah, 1987 Cancer Res. 47:5924-5931; Wawrzynczak et ah, In Immunnoconjugates: Antibody Conjugates in Radioimmagery and Therapy of Cancer, C.W.Vogel ed, Oxford U. Press, 1987; U.S. Patent No. 4,880,935). The linker can be a malonate linker (Johnson et al, 1995, Anticancer Res. 15: 1387-93), a maleimidobenzoly linker (lau et al, 1995, Bioorg- Med-Chem. 3(10):1299-1304) or a 3'-N-amide analog (Lau et al, 1995, Bioorg-Med- Chem.3(10):1305-1312). Prophylactic and Therapeutic Uses of the Present Invention
The antibodies of the invention can act as either agonists or antagonists of GPNMB, depending on the methods of their use. The antibodies can be used to prevent, diagnose, or treat medical disorders in a subject, especially in humans. Antibodies of the invention can also be used for isolating GPNMB or GPNMB-expi'essing cells. Furthermore, the antibodies can be used to treat a subject at risk of or susceptible to a disorder or having a disorder associated with aberrant GPNMB expression or function. Antibodies of the invention can be used to detect GPNMB in such subjects.
The present invention provides methods for treating and/or preventing a disease or disorder associated with overexpression of GPNMB and/or cell hyperproliferative disorders, particularly cancer, in a subject comprising administering an effective amount of a composition that can target cells expressing GPNMB, and inhibiting the GPNMB expression or function, and/or having therapeutic or prophylactic effects on the hyperproliferative cell disease. In one embodiment, the method of the invention comprises administering to a subject a composition comprising an immunoconjugate that comprises an antibody of the invention and a cytotoxic agent against the hyperproliferative cell disease. In another embodiment, the method of the invention comprises administering to a subject in need thereof a composition comprising a naked IgGl antibody of the invention and one or more immunomodulators. In yet another embodiment, the method of the invention comprises administering to a subject in need thereof a composition comprosing a single chain Fv antibody (anti-GPNMB) conjugated to a cytotoxic agent, or a composition comprising a bispecific antibody that have a single chain anti-GPNMB antibody component and a anti-CD3 antibody component. In a preferred embodiment, the hyperproliferative cell disease is cancer. More preferably, the cancer is melanoma, or a cancer of the CNS system, such as astrocytoma, glioblastoma, medulloblastoma, or neoplastic meningitis. The present invention provides therapies comprising administering one of more antibodies of the invention and compositions comprising said antibodies to a subject, preferably a human subject, for preventing and/or treating a disorder characterized by or associated with aberrant expression and/or activity of GPNMB or a symptom thereof. In one embodiment, the invention provides a method of preventing or treating a disorder characterized by or associated with aberrant expression and/or activity of GPNMB or a symptom thereof, said method comprising administering to a subject in need thereof an effective amount of one or more antibodies of the invention. In certain embodiments, an effective amount of one or more immunoconjugates comprising one or more antibodies of the invention is administered to a subject in need thereof to prevent or treat a disorder characterized by or associated with aberrant expression and/or activity of GPNMB or a symptom thereof.
The invention also provides methods of preventing or treating a disorder characterized by or associated with aberrant expression and/or activity of GPNMB or a symptom thereof, said methods comprising administering to a subject in need thereof one or more of the antibodies of the invention and one or more therapies (e.g., one or more prophylactic or therapeutic agents) other than antibodies of the invention. The prophylactic or therapeutic agents of the combination therapies of the invention can be administered sequentially or concurrently. In a specific embodiment, the combination therapies of the invention comprise an effective amount of one or more antibodies of the invention and an effective amount of at least one other therapy (e.g., prophylactic or therapeutic agent) which has a different mechanism of action than said antibodies. In certain embodiments, the combination therapies of the present invention improve the prophylactic or therapeutic effect of one or more antibodies of the invention by functioning together with the antibodies to have an additive or synergistic effect. In certain embodiments, the combination therapies of the present invention reduce the side effects associated with the therapies (e.g., prophylactic or therapeutic agents).
The prophylactic or therapeutic agents of the combination therapies can be administered to a subject, preferably a human subject, in the same pharmaceutical composition. Alternatively, the prophylactic or therapeutic agents of the combination therapies can be administered concurrently to a subject in separate pharmaceutical compositions. The prophylactic or therapeutic agents may be administered to a subject by the same or different routes of administration. In a specific embodiment, a pharmaceutical composition comprising one or more antibodies of the invention described herein is administered to a subject, preferably a human, to prevent and/or treat a disorder characterized by or associated with aberrant expression and/or activity of GPNMB or a symptom thereof. In accordance with the invention, pharmaceutical compositions of the invention may also comprise one or more therapies (e.g., prophylactic or therapeutic agents), other than antibodies of the invention.
The antibodies of the invention may also be used to detect the presence of GPNMB in biological samples (in diagnostic methods or use as an efficacy marker). The amount of GPNMB detected may be correlated with the expression level of GPNMB, which, in turn, is correlated with the disease, tumor type, tumor burden or stage using methods known in the art (see for example recommendations of the AAPS Ligand Binding Assay Bioanalytical Focus Group (LBABFG) Pharm Res. 2003 Nov;20(ll): 1885-900). Detection methods that employ antibodies are well known in the art and include, for example, ELISA, radioimmunoassay, immunoblot, Western blot, IHC, immunofluorescence, immunoprecipitation. The antibodies may be provided in a diagnostic kit that incorporates one or more of these techniques to detect GPNMB. Such a kit may contain other components, packaging, instructions, or other material to aid the detection of the protein. In a specific embodiment, the antibodies of the invention are conjugated to a radioactive isotope, and are injected to a subject to detect cells that overexpressing GPNMB.
Where the antibodies are intended for diagnostic purposes, it may be desirable to modify them, for example, with a ligand group (such as biotin) or a detectable marker group (such as a fluorescent group, a radioisotope or an enzyme). If desired, the antibodies of the invention may be labeled using conventional techniques. Suitable detectable labels include, for example, fiuorophores, chromophores, radioactive atoms, electron-dense reagents, enzymes, and ligands having specific binding partners. Enzymes are typically detected by their activity. For example, horseradish peroxidase can be detected by its ability to convert tetramethylbenzidine (TMB) to a blue pigment, quantifiable with a spectrophotometer. For detection, suitable binding partners include, but are not limited to, biotin and avidin or streptavidin, IgG and protein A, and the numerous receptor-ligand couples known in the art. Other permutations and possibilities will be readily apparent to those of ordinary skill in the art, and are considered as equivalents within the scope of the instant invention.
Antibodies of the invention can be used in screening methods to identify inhibitors of GPNMB effective as therapeutics. In such a screening assay, a first binding mixture is formed by combining GPNMB and an antibody of the invention; and the amount of binding in the first binding mixture (M0) is measured. A second binding mixture is also formed by combining GPNMB, the antibody, and the compound or agent to be screened, and the amount of binding in the second binding mixture (Mi) is measured. A compound to be tested may be another anti-GPNMB antibody. The amounts of binding in the first and second binding mixtures are then compared, for example, by calculating the Mi /Mo ratio. The compound or agent is considered to be capable of modulating a GPNMB-associated responses if a decrease in binding in the second binding mixture as compared to the first binding mixture is observed. The formulation and optimization of binding mixtures is within the level of skill in the art, such binding mixtures may also contain buffers and salts necessary to enhance or to optimize binding, and additional control assays may be included in the screening assay of the invention. Compounds found to reduce the GPNMB-antibody binding by at least about 10% {i.e., Mj/M 00.9), preferably greater than about 30% may thus be identified and then, if desired, secondarily screened for the capacity to ameliorate a disorder in other assays or animal models as described below. The strength of the binding between GPNMB and an antibody can be measured using, for example, an enzyme-linked immunoadsorption assay (ELISA), radioimmunoassay (RIA), surface plasmon resonance- based technology (e.g., Biacore), all of which are techniques well known in the art. The compound may then be tested in vitro as described in the Examples, infra.
Dosage and Frequency of Administration
The amount of a prophylactic or therapeutic agent or a composition of the invention which will be effective in the prevention and/or treatment of a disorder associated with or characterized by aberrant expression and/or activity of GPNMB can be determined by standard clinical methods. For example, the dosage of the composition which will be effective in the treatment and/or prevention of cancer can be determined by administering the composition to an animal model. In addition, in vitro assays may optionally be employed to help identify optimal dosage ranges. Preliminary doses as, for example, determined according to animal tests, and the scaling of dosages for human administration is performed according to art-accepted practices. Toxicity and therapeutic efficacy can be determined by standard pharmaceutical procedures in cell cultures or experimental animals. The data obtained from the cell culture assays or animal studies can be used in formulating a range of dosage for use in humans. Therapeutically effective dosages achieved in one animal model can be converted for use in another animal, including humans, using conversion factors known in the art (see, e.g., Freireich et al. (1966) Cancer Chemother. Reports, 50(4): 219-244).
Selection of the preferred effective dose can be determined (e.g., via clinical trials) by a skilled artisan based upon the consideration of several factors which will be known to one of ordinary skill in the art. Such factors include the disease to be treated or prevented, the symptoms involved, the patient's body mass, gender, immune status and other factors known by the skilled artisan to reflect the accuracy of administered pharmaceutical compositions. Suitable regimens can be selected by one skilled in the art by considering such factors and by following, for example, dosages reported in literature and recommended in the Physician's Desk Reference (59th ed., 2005).
The precise dose to be employed in the formulation will also depend on the route of administration, and the seriousness of the cancer, and should be decided according to the judgment of the practitioner and each patient's circumstances. Effective doses may be extrapolated from dose-response curves derived from in vitro or animal model test systems.
For other cancer therapeutic agents administered to a patient, the typical doses of various cancer therapeutics are known in the art. Given the invention, certain preferred embodiments will encompass the administration of lower dosages in combination treatment regimens than dosages recommended for the administration of single agents. In a specific embodiment, the dosage of an antibody or an immunoconjugate comprising an antibody of the invention administered to prevent and/or treat a disorder associated with or characterized by aberrant expression and/or activity of GPNMB {e.g., cancer) in a patient is 30 mg/kg or less, 25 mg/kg or less, 20 mg/kg or less, 15 nig/kg or less, preferably 12 mg/kg or less, 11 mg/kg or less, 10 mg/kg or less, 9 mg/kg or less, 8 mg/kg or less, 7 mg/kg or less, 6 mg/kg or less, 5 mg/kg or less, 4 mg/kg or less, 3 mg/kg or less, 2 mg/kg or less, or 1 mg/kg or less of a patient's body weight. In another embodiment, the dosage of an antibody or an immunoconjugate of the invention administered to prevent and/or treat a disorder associated with or characterized by aberrant expression and/or activity of GPNMB {e.g., cancer) in a patient is a unit dose of about 0.01 mg/kg to about 20 mg/kg, about 0.1 mg/kg to about 10 mg/kg, about 0.1 mg/kg to about 8 mg/kg, about 0.1 mg/kg to about 7 mg/kg, about 0.1 mg/kg to about 6 mg/kg, about 0.1 mg/kg to about 5 mg/kg, about 0.1 mg/kg to about 4 mg/kg, preferably, about 0.1 mg/kg to about 3 mg/kg, about 0.2 mg/kg to 3 mg/kg, about 0.3 mg/kg to about 3 mg/kg, about 0.4 mg/kg to about 3 mg/kg, about 0.6 mg/kg to about 3 mg/kg, about 0.8 mg/kg to about 3 mg/kg, about 0.1 mg/kg to 2 mg/kg, about 0.1 mg/kg to 1 mg/kg. In certain embodiments, the dosage of an antibody or an immunoconjugate comprising an antibody of the invention administered to prevent and/or treat a disorder associated with or characterized by aberrant expression and/or activity of GPNMB {e.g., cancer) in a patient is a unit dose of about 0.1 mg/kg, about 0.2 mg/kg, about 0.4 mg/kg, about 0.6 mg/kg, about 0.8 mg/kg, about 1.1 mg/kg, or about 1 mg/kg.
In certain embodiments, a subject is administered one or more doses of an effective amount of one or more antibodies or imunoconjugates of the invention to prevent and/or treat a disorder associated with or characterized by aberrant expression and/or activity of GPNMB, wherein the dose of an effective amount of said antibodies, immunoconjugates, compositions, or combination therapies reduces and/or inhibits proliferation of cancerous cells by at least 20% to 25%, preferably at least 25% to 30%, at least 30% to 35%, at least 35% to 40%, at least 40% to 45%, at least 45% to 50%, at least 50% to 55%, at least 55% to 60%, at least 60% to 65%, at least 65% to 70%, at least 70% to 75%, at least 75% to 80%, at least 80 to 85%, at least 85% to 90%, at least 90% to 95%, or at least 95% to 98% relative to a control such as PBS in an in vitro and/or in vivo assay well-known in the art.
In other embodiments, a subject is administered one or more doses of an effective amount of one or more antibodies or immunoconjugates of the invention to prevent and/or treat a disorder associated with or characterized by aberrant expression and/or activity of GPNMB, wherein the dose of an effective amount achieves a serum titer of at least 0.1 μg/mL, at least 0.5 μg/mL, at least 1 μg/mL, at least 2 μg/mL, at least 5 μg/mL, at least 6 μg/mL, at least 10 μg/mL, at least 15 μg/mL, at least 20 μg/mL, at least 25 μg/mL, at least 50 μg/mL, at least 100 μg/mL, at least 125 μg/mL, at least 150 μg/mL, at least 175 μg/mL, at least 200 μg/mL, at least 225 μg/mL, at least 250 μg/mL, at least 275 μg/mL, at least 300 μg/mL, at least 325 μg/mL, at least 350 μg/mL, at least 375 μg/mL, or at least 400 μg/mL of the antibodies of the invention. In yet other embodiments, a subject is administered a dose of an effective amount of one or more antibodies or immunoconjugates of the invention to achieve a serum titer of at least 0.1 μg/mL, at least 0.5 μg/mL, at least 1 μg/mL, at least, 2 μg/mL, at least 5 μg/mL, at least 6 μg/mL, at least 10 μg/mL, at least 15 μg/mL, at least 20 μg/mL, at least 25 μg/mL, at least 50 μg/mL, at least 100 μg/mL, at least 125 μg/mL, at least 150 μg/mL, at least 175 μg/mL, at least 200 μg/mL, at least 225 μg/mL, at least 250 μg/mL, at least 275 μg/mL, at least 300 μg/mL, at least 325 μg/mL, at least 350 μg/mL, at least 375 μg/mL, or at least 400 μg/mL of the antibodies and a subsequent dose of an effective amount of one or more antibodies or imunoconjugates of the invention is administered to maintain a serum titer of at least 0.1 μg/mL, at least 0.5 μg/mL, at least 1 μg/mL, at least, 2 μg/mL, at least 5 μg/mL, at least 6 μg/mL, at least 10 μg/mL, at least 15 μg/mL, at least 20 μg/mL, at least 25 μg/mL, at least 50 μg/mL, at least 100 μg/mL, at least 125 μg/mL, at least 150 μg/mL, at least 175 μg/mL, at least 200 μg/mL, at least 225 μg/mL, at least 250 μg/mL, at least 275 μg/mL, at least 300 μg/mL, at least 325 μg/mL, at least 350 μg/mL, at least 375 μg/mL, or at least 400 μg/mL. In accordance with these embodiments, a subject may be administered 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 or more subsequent doses. In a specific embodiment, the invention provides methods of preventing and/or treating a disorder associated with or characterized by aberrant expression and/or activity of GPNMB, said method comprising administering to a subject in need thereof a unit dose of at least O.Olmg/kg, at least 0.1mg/kg, at least 0.2mg/kg, at least 0.4mg/kg, at least 0.6mg/kg, at least 0.8mg/kg, at least lmg/kg, or at least 1.lmg/kg of one or more antibodies or immunoconjugates of the invention. In another embodiment, the invention provides methods of preventing and/or treating a disorder associated with or characterized by aberrant expression and/or activity of GPNMB, said method comprising administering to a subject in need thereof a unit dose of at least 0.01 mg/kg, at least 0.1 mg/kg, at least 0.2 mg/kg, at least 0.4 mg/kg, at least 0.6 mg/kg, at least 0.8 mg/kg, at least 1 mg/kg, or at least 1.1 mg/kg of one or more antibodies or immunoconjugates of the invention once every 7 days, preferably, once every 10 days, once every 12 days, once every 14 days, once every 16 days, once every 18 days, once every three weeks, or once a month. In a preferred embodiment, an immunoconjuage of the instant invention is administered at a unit dose of about 0.1 mg/kg, about 0.2 mg/kg, about 0.4 mg/kg, about 0.6 mg/kg, about 0.8 mg/kg, about 1.1 mg/kg, or about 1 mg/kg once every 10 to 20 days with 2 to 4 cycles.
The present invention provides methods of preventing and/or treating a disorder associated with or characterized by aberrant expression and/or activity of GPNMB, said method comprising: (a) adtninistering to a subject in need thereof one or more doses of a prophylactically or therapeutically effective amount of one or more antibodies or immunoconjugates of the invention; and (b) monitoring the plasma level/concentration of the said administered antibody or antibodies in said subject after administration of a certain number of doses of the said antibody or antibodies. Moreover, preferably, said certain number of doses is 1, 2, 3, 4, 5, 6, 7, or 8 doses of a prophylactically or therapeutically effective amount one or more antibodies or immunoconjugates of the invention. In a specific embodiment, the invention provides a method of preventing and/or treating a disorder associated with or characterized by aberrant expression and/or activity of GPNMB, said method comprising: (a) administering to a subject in need thereof a dose of at least 0.1 mg/kg (preferably at least at least 0.2 mg/kg, at least 0.4 mg/kg, at least 0.6 mg/kg, at least 0.8 mg/kg, at least 1 mg/kg, or at least 1.1 mg/kg) of one or more antibodies or iminunoconjugates of the invention; and (b) administering one or more subsequent doses to said subject when the plasma level of the antibody or antibodies administered in said subject is less than 0.1 μg/mL, preferably less than 0.25 μg/mL, less than 0.5 μg/mL, less than 0.75 μg/mL, or less than 1 μg/mL. In another embodiment, the invention provides a method of preventing and/or treating a disorder associated with or characterized by aberrant expression and/or activity of GPNMB, said method comprising: (a) administering to a subject in need thereof one or more doses of at least at least 0.1 mg/kg (preferably at least at least 0.2 mg/kg, at least 0.4 mg/kg, at least 0.6 mg/kg, at least 0.8 mg/kg, at least 1 mg/kg, or at least 1.1 mg/kg) of one or more antibodies of the invention; (b) monitoring the plasma level of the administered antibody or antibodies of the invention in said subject after the administration of a certain number of doses; and (c) administering a subsequent dose of the antibody or antibodies of the invention when the plasma level of the administered antibody or antibodies in said subject is less than 0.1 μg/mL, preferably less than 0.25 μg/mL, less than 0.5 μg/mL, less than 0.75 μg/mL, or less than 1 μg/mL. Preferably, said certain number of doses is 1, 2, 3, 4, 5, 6, 7, or 8 doses of an effective amount of one or more antibodies or immunoconjugates of the invention.
Therapies (e.g., prophylactic or therapeutic agents), other than antibodies or immunoconjugates of the invention, which have been or are currently being used to prevent and/or treat a disorder associated with or characterized by aberrant expression and/or activity of GPNMB can be administered in combination with one or more antibodies or immunoconjugates of the invention according to the methods of the invention to treat and/or prevent a disorder associated with or characterized by aberrant expression and/or activity of GPNMB. Preferably, the dosages of prophylactic or therapeutic agents used in combination therapies of the invention are lower than those which have been or are currently being used to prevent and/or treat a disorder associated with or characterized by aberrant expression and/or activity of GPNMB.
In various embodiments, the therapies (e.g., prophylactic or therapeutic agents) are administered less than 5 minutes apart, less than 30 minutes apart, 1 hour apart, at about 1 hour apart, at about 1 to about 2 hours apart, at about 2 hours to about 3 hours apart, at about 3 hours to about 4 hours apart, at about 4 hours to about 5 hours apart, at about 5 hours to about 6 hours apart, at about 6 hours to about 7 hours apart, at about 7 hours to about 8 hours apart, at about 8 hours to about 9 hours apart, at about 9 hours to about 10 hours apart, at about 10 hours to about 1 1 hours apart, at about 11 hours to about 12 hours apart, at about 12 hours to 18 hours apart, 18 hours to 24 hours apart, 24 hours to 36 hours apart, 36 hours to 48 hours apart, 48 hours to 52 hours apart, 52 hours to 60 hours apart, 60 hours to 72 hours apart, 72 hours to 84 hours apart, 84 hours to 96 hours apart, or 96 hours to 120 hours part. In preferred embodiments, two or more therapies are administered within the same patient visit.
In certain embodiments, one or more antibodies of the invention and one or more other therapies (e.g., prophylactic or therapeutic agents) are cyclically administered. Cycling therapy involves the administration of a first therapy (e.g., a first prophylactic or therapeutic agent) for a period of time, followed by the administration of a second therapy (e.g., a second prophylactic or therapeutic agent) for a period of time, optionally, followed by the administration of a third therapy (e.g., prophylactic or therapeutic agent) for a period of time and so forth, and repeating this sequential administration, i.e., the cycle in order to reduce the development of resistance to one of the therapies, to avoid or reduce the side effects of one of the therapies, and/or to improve the efficacy of the therapies. Pharmaceutical Compositions and Methods of Administration
The disclosure provides compositions comprising anti-GPNMB antibodies. Such compositions may be suitable for pharmaceutical use and administration to patients. The compositions typically comprise one or more antibodies of the present invention and a pharmaceutically acceptable excipient. The phrase "pharmaceutically acceptable excipient" includes any and all solvents, dispersion media, coatings, antibacterial agents and antifungal agents, isotonic agents, and absorption delaying agents, and the like, that are compatible with pharmaceutical administration. The use of such media and agents for pharmaceutically active substances is well known in the art. The compositions may also contain other active compounds providing supplemental, additional, or enhanced therapeutic functions. The pharmaceutical compositions may also be included in a container, pack, or dispenser together with instructions for administration.
A pharmaceutical composition of the invention is formulated to be compatible with its intended route of administration. Methods to accomplish the administration are known to those of ordinary skill in the art. The administration may, for example, be intravenous, intraperitoneal, intramuscular, intracavity, subcutaneous or transdermal. It may also be possible to obtain compositions which may be topically or orally administered, or which may be capable of transmission across mucous membranes. Solutions or suspensions used for intradermal or subcutaneous application typically include one or more of the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerin, propylene glycol, or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates; and agents for the adjustment of tonicity such as sodium chloride or dextrose. The pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide. Such preparations may be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic. Pharmaceutical compositions suitable for injection include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion. For intravenous administration, suitable carriers include physiological saline, bacteriostatic water, Cremophor EL (BASF, Parsippany, NJ.) or phosphate buffered saline (PBS). In all cases, the composition must be sterile and should be fluid to the extent that easy syringability exists. It should be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi. Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars; polyalcohols such as mannitol, sorbitol, and sodium chloride in the composition. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyetheylene glycol, and the like), and suitable mixtures thereof. The proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and/or by the use of surfactants. Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent which delays absorption, for example, aluminum monostearate, and gelatin.
Oral compositions generally include an inert diluent or an edible carrier. They can be enclosed in gelatin capsules or compressed into tablets. For oral administration, the antibodies can be combined with excipients and used in the form of tablets, troches, or capsules. Pharmaceutically compatible binding agents, and/or adjuvant materials can be included as part of the composition. The tablets, pills, capsules, troches, and the like can contain any of the following ingredients, or compounds of a similar nature; a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or com starch; a lubricant such as magnesium stearate or Sterotes; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring.
Systemic administration can also be by transmucosal or transdermal means. For transmucosal or transdermal administration, penetrants appropriate to the harrier to be permeated are used in the formulation. Such penetrants are generally known in the art, and include, for example, detergents, bile salts, and fusidic acid derivatives. Transmucosal administration may be accomplished, for example, through the use of lozenges, nasal sprays, inhalers, or suppositories. For example, in case of antihodies that comprise the Fc portion, compositions may be capable of transmission across mucous membranes in intestine, mouth, or lungs (e.g., via the FcRn receptor-mediated pathway as described in U.S. Pat. No. 6,030,613). For transdermal administration, the active compounds may be formulated into ointments, salves, gels, or creams as generally known in the art. For administration by inhalation, the antibodies may be delivered in the form of an aerosol spray from pressured container or dispenser, which contains a suitable propellant, e.g., a gas such as carbon dioxide, or a nebulizer. In certain embodiments, the presently disclosed antibodies are prepared with carriers that will protect the compound against rapid elimination from the body, such as a controlled release formulation, including implants and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Methods for preparation of such formulations will be apparent to those skilled in the art. Liposomal suspensions containing the presently disclosed antibodies can also be used as pharmaceutically acceptable carriers. These can be prepared according to methods known to those skilled in the art, for example, as described in U.S. Pat. No. 4,522,811.
It may be advantageous to formulate oral or parenteral compositions in a dosage unit form for ease of administration and uniformity of dosage. The term "dosage unit form" as used herein refers to physically discrete units suited as unitary dosages for the subject to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
Toxicity and therapeutic efficacy of the composition of the invention can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD5O (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD50/ED50. Compositions that exhibit large therapeutic indices are preferred.
For any composition used in the present invention, the therapeutically effective dose can be estimated initially from cell culture assays. Examples of suitable bioassays include DNA replication assays, clonogenic assays and other assays as, for example, described in the Examples. The data obtained from the cell culture assays and animal studies can be used in formulating a range of dosage for use in humans. A dose may be formulated in animal models to achieve a circulating plasma concentration range that includes the IC50 (i.e., the concentration of the antibody which achieves a half-maximal inhibition of symptoms). Circulating levels in plasma may be measured, for example, by high performance liquid chromatography. The effects of any particular dosage can be monitored by a suitable bioassay. The dosage lies preferably within a range of circulating concentrations with little or no toxicity. The dosage may vary depending upon the dosage form employed and the route of administration utilized.
Antibodies can be modified to become immunotoxins utilizing techniques that are well known in the art (Vitetta 1993, Immunol Today 14:252; U.S. Patent No. 5,194,594). Cyotoxic immunoconjugates are known in the art and have been used as therapeutic agents. Such immunoconjugates may for example, use maytansinoids (US 6,441,163), tubulin polymerization inhibitor, auristatin (Mohammad et al, 1999 Int. J. Oncol 15(2):367-72; Dorotήna et al, 2003 Nature Biotechnology 21(7): 778-784), dolastatin derivatives (Ogawa et al, 2001 Toxicol Lett. 121(2):97-106) 21(3)778- 784), Mylotarg® (Wyeth Laboratories, Philidelphia, PA); maytansinoids (DMl), taxane or mertansine (ImmunoGen Inc.).
Immunoradiopharmaceuticals utilizing anti-GPNMB antibodies may be prepared utilizing techniques mat are well known in the art (Junghans et al. in Cancer Chemotherapy and Biothβrapy 655-686 (2d edition, Chafner and Longo, eds., Lippincott Raven (1996);US. Patent Nos. 4,681,581, 4, 735,210, 5,101,827, 5,102,990 (RE 35,500), 5,648,471, and 5,697,902). Each of the immunotoxins and radiolabeled antibody molecules selectively kill cells expressing GPNMB. Radiolabels are known in the art and have been used for diagnostic or therapeutic radioimmuno conjugates. Examples of radiolabels include, but are not limited to, the following: radioisotopes or radionuclides (e.g., 3H, 14C, 15N, 35S, 90Y, 99Tc, 111In, 125I, 131I, 177Lu, 105Rh, Rhenium-186, Rhenium-188, Samarium- 153, Copper-64, Scandium-47). For example, radionuclides which have been used in radioimmunoconjugate guided clinical diagnosis include, but are not limited to: 131 I, 125 1, 123 j^ 99 ^ 67 Qa^ ag wejj ag i n jn Antibodies have also been labeled with a variety of radionuclides for potential use in targeted immunotherapy (see Peirersz et ah, 1987). These radionuclides include, for example, ' * Re and ' Re as well as 90 Y, and to a lesser extent ' " Au and bl Cu. I-( 131 ) {see for example U. S. Pat. No. 5, 460, 785) . Radio therapeutic chelators and chelator conjugates are known in the art (U.S. 4,831,175, 5,099,069, 5,246,692, 5,286,850, and 5,124,471).
EXAMPLES The following examples, including the experiments conducted and results achieved are provided for illustrative purposes only and are not to be construed as limiting upon the present invention. Example 1: Immunogen
Recombinant human GPNMB (SEQ ID NO:289), specifically the extra-cellular domain (ECD) was prepared for use as the immunogen. Generally, cDNA encoding the ECD of GPNMB with a C-terminus V5-HIS tag was transfected into HEK 293 cells, expressed and purified using cation exchange chromatography with a POROS HS 50 (Applied Biosystems, Foster City, CA). Sample was eluted with IM NaCl at a pH of 5.5, followed by metal affinity chromatography (Pharmacia metal chelate 5 mL). The sample was eluted against a linear gradient from 10-500 mM imidazole over 10 CV (column volumn). Dialysis occurred using 2OmM Tris/ 5OmM NaCl at pH 7.4 (2L x 2). The sample was then filtered through a 0.22 μm filter.
Example 2: Immunization A preferred method for generating fully human antibodies uses XenoMouse© strains of mice which have been engineered to contain 245 kb and 190 kb-sized germline configuration fragments of the human heavy chain locus and kappa light chain locus (Green et al. 1994 Nature Genetics 7:13-21; Mendez et al. 1997 Nature Genetics 15:146-156; Green and Jakobovits, 1998 J. Exp. Med. 188:483-495; U.S. Patent Nos. 6, 162,963, 6,150,584, 6,114,598, 6,075, 181, and 5,939,598.) In an alternative approach, the minilocus approach, an exogenous Ig locus is mimicked through the inclusion of pieces (individual genes) from the Ig locus. Thus, one or more VH genes, one or more DH genes, one or more JH genes, a mu constant region, and a second constant region (preferably a gamma constant region) are formed into a construct for insertion into an animal (Taylor et al, 1992, Chen et al, 1993, Tuaillon et al, 1993, Choi et al, 1993, Lonherg et al, (1994), Taylor et al, (1994), and Tuaillon et al., (1995), Fishwild et al, (1996); U.S. Patent Nos. 5,545,807, 5,545,806, 5,625,825, 5,625,126, 5,633,425, 5, 661,016, 5, 770,429, 5, 789,650, 5,814,318, 5,877,397, 5,874,299, 6,255,458, 5,591,669, 6,023,010, 5,612,205, 5, 721,367, 5, 789,215, 5, 643, 763, 5,981, 175). It is understood that the λK XenoMouse® may be used to generate anti-GPNMB antibodies utilizing lambda V regions. Such antibodies are within the scope of the invention.
Immunization GPNMB-V5His immunogen (as prepared in Example 1) was used as an antigen.
Monoclonal antibodies against GPNMB were developed by sequentially immunizing XenoMouse® mice (XenoMouse' XMG2 strain), Abgenix, Inc. Fremont, CA. XenoMouse® animals were immunized via footpad route for all injections. The total volume of each injection was 50 μl per mouse, 25 μl per footpad. For cohort 1 (10 XMG2 mice), the initial immunization was with 10 μg of
GPNMB-V5His admixed 1 :1 (v/v) with 100 μg alum gel ("Adju-Phos": aluminum phosphate gel adjuvant, Superfos BIOSECTOR™ a/s, distributed by E.M. Sergent Pulp and Chemical Co., Clifton, NJ, cat. # 1452-250) per mouse. The subsequent five boosts were made with 5 μg of GPNMB-V5His admixed 1:1 (v/v) with 100 μg alum gel in pyrogen-free D-PBS. The seventh boost consisted of 5 μg of GPNMB-V5His admixed 1:1 (v/v) with TITERMAX GOLD®(Sigma; cat. # T2684). The eighth injection consisted of 5 μg of GPNMB -V5His admixed 1 :1 v/v with 100 μg alum gel . A final boost was made with 5 μg GPNMB-V5His in pyrogen-free DPBS, without adjuvant. The XenoMouse® mice were immunized on days 0, 3, 6, 10, 14, 17, 23, and 27 for this protocol and fusions were performed on day 31. The bleed was made through Retro-Orbital Bleed procedure on day 21 after the sixth boost . For cohort 2 (10 XMG2 mice), the initial immunization was with 10 μg of
GPNMB-V5His admixed 1:1 (v/v) with 100 μg alum gel per mouse. The subsequent two boosts were made with 5 μg of GPNMB-V5His admixed 1 :1 (v/v) with 100 μg alum gel in pyrogen-free D-PBS. The fourth boost consisted of 5 μg of GPNMB-V5His admixed 1:1 (v/v) with TITERMAX GOLD®(Sigma; cat. # T2684). The following fifth to seventh injection consisted of 5 μg of GPNMB-V5His admixed 1:1 v/v with 100 μg alum gel . The eighth injection and final boost was made with 5 μg GPNMB-V5His in pyrogen-free
DPBS, without adjuvant. The XenoMouse® mice were immunized on days 0, 3, 7, 11, 14,
17, 22, 25 and 74. for this protocol and fusions were performed on day 78. The bleeds was made through Retro-Orbital Bleed procedure on day 21 after the sixth boost.
The footpad injection was performed by the following protocol using only the ventral surface of both hind limb paws. A solution was injected beneath the skin without piercing the muscle tissue by using an insulin 1/2 mL syringe with attached 28 or 30 gauge x 1/2" needle. The mouse to be injected was grasped by the loose fur along its neck and back so that it was immobilized and was turned over so the ventral side was accessible. The hind limb of the mouse was grasped and the needle was inserted (bevel side up) at the ankle, threading just under the skin until the needle tip reached the paw. The needle was inserted along the outside length of the hind foot carefully, to avoid the vein located towards the inner side of the foot. Once the tip of the needle reached the paw, the solution was injected slowly until resistance was felt or the designated volume had been dispensed. The needle was then withdrawn and the second hind foot injected in the same manner.
The following Table 4 provides the immunization schedule for the 2 groups of mice. Table 4:Immunization Schedule of GPNMB Anti en: GPNMB-soluble at 043m /mL
Figure imgf000058_0001
Figure imgf000058_0002
Figure imgf000059_0001
Figure imgf000059_0002
Selection of animals for harvest by titer
Anti- GPNMB antibody titers in the serum from immunized XenoMouse® mice were determined by ELISA. Briefly, three sets of ELISAs were set up. GPNMB (+NMB) at 1 μg/mL, GPNMB(-NMB) at 1 μg/mL, and NMB at 1 μg/mL were coated onto Costar Labcoat Universal Binding Polystyrene 96-well plates (Corning, Acton, MA) overnight at 40C in Antigen Coating Buffer (0.1 M Carbonate Buffer, pH 9.6 NaHCO3 (MW 84) 8.4 g/L). The next day, the plates were washed three times with washing buffer (0.05% Tween 20 in Ix PBS) using a Biotek plate washer. The plates were then blocked with 200 ul/well blocking buffer (0.5% BSA, 0.1% Tween 20, 0.01% Thimerosal in Ix PBS) and incubated at room temperature for 1 h. After the one-hour blocking, the plates were washed three times with washing buffer using a Biotek plate washer. Sera from either GPNMB immunized XenoMouse® mice or naϊve XenoMouse® animals were titrated in 0.5% BSA/PBS buffer at 1:3 dilutions in duplicate from a 1:100 initial dilution. The last well was left blank. These plates were incubated at room temperature for 2 h, and the plates were then washed three times with washing buffer using a Biotek plate washer. A goat anti- human IgG Fc-specific horseradish peroxidase (HRP, Pierce, Rockford, IL) conjugated antibody was added at a final concentration of 1 μg/mL and incubated for 1 hour at room temperature. The plates were washed three times with washing buffer using a Biotek plate washer. After washing, the plates were developed with the addition of TMB chromogenic substrate (BioFx BSTP-0100-01) for 10-20 min or until negative control wells start to show color. Then the ELISA was stopped by the addition of Stop Solution (650 iiM Stop reagent for TMB (BioFx BSTP-0100-01), reconstituted with 100 mL H2O per bottle). The specific titer of each XenoMouse® animal was determined from the optical density at 650 nm and is shown in Tables 2 and 3 below. The titer value is the reciprocal of the greatest dilution of sera with an OD reading two-fold that of background. Therefore, the higher the number, the greater was the humoral immune response to GPNMB. The results are provided in Table 5. Table 5: XENOMOUSE® Anti-GPNMB Serum titers
Figure imgf000060_0001
Pooled anti-GPNMB sera from immunized animals was also evaluated by FACS for reactivity to UACC-62, SF539, SKMEL5, U87MG, and LOXlMVI cell lines. Pooled sera were tested at 1:10, 1:100 and 1:500 compared to Anti-IL13 serum (control) and prebleeds diluted at 1 : 10, 1 :100 (control). Example 3: Antibodies
Hybridoma cell lines were generated from immunized mice demonstrated to have anti-GPNMB titers using standard techniques (see Mendez et al, 1997, Nat Genet. 15: 146- 156).
Immunized mice were sacrificed by cervical dislocation, and the lymph nodes were harvested and pooled from each cohort. The lymphoid cells were dissociated by grinding in
DMEM to release the cells from the tissues, and the cells were suspended in DMEM. The cells were counted, and 0.9 mL DMEM per 100 million lymphocytes was added to the cell pellet to resuspend the cells gently but completely. Using 100 μl of CD90+ magnetic beads per 100 million cells, the cells were labeled by incubating the cells with the magnetic beads at 4°C for 15 minutes. The magnetically-labeled cell suspension containing up to 108 positive cells (or up to 2x109 total cells) was loaded onto a LS+ column and the column washed with DMEM. The total effluent was collected as the CD90-negative fraction (most of these cells were expected to be B cells).
The fusion was performed by mixing washed enriched B cells from above and nonsecretory myeloma P3X63Ag8.653 cells purchased from ATCC, cat.# CRL 1580
(Kearney et al, J. Immunol. 123, 1979, 1548-1550) at a ratio of 1:1. The cell mixture was gently pelleted by centrifugation at 800 g. After complete removal of the supernatant, the cells were treated with 2-4 mL of Pronase solution (CalBiochem, cat. # 53702; 0.5 mg/mL in PBS) for no more than 2 minutes. Then 3-5 mL of FBS was added to stop the enzyme activity and the suspension was adjusted to 40 mL total volume using electro cell fusion solution, ECFS (0.3 M Sucrose, Sigma, Cat# S7903, 0.1 mM Magnesium Acetate, Sigma,
Cat# M2545, 0.1 mM Calcium Acetate, Sigma, Cat# C4705). The supernatant was removed after centrifugation and the cells were resuspended in 40 mL ECFS. This wash step was repeated and the cells again were resuspended in ECFS to a concentration of 2x10δ cells/mL.
Electro-cell fusion was performed using a fusion generator, model ECM2001, Genetronic, Inc., San Diego, CA. The fusion chamber size used was 2.0 mL, using the Abgenix, Inc. optimum instrument settings to do ECF. After ECF, the cell suspensions were carefully removed from the fusion chamber under sterile conditions and transferred into a sterile tube containing the same volume of Hybridoma Culture Medium (DMEM (JRH Biosciences), 15% FBS (Hyclone), supplemented with L-glutamine, pen/strep, OPI (oxaloacetate, pyruvate, bovine insulin) (all from Sigma) and IL-6 (Boehringer Mannheim)). The cells were incubated for 15-30 minutes at 370C, and then centrifuged at 400 g (1000 rpm [but in what rotor? Otherwise, leave out the rpm]) for five minutes. The cells were gently resuspended in a small volume of Hybridoma Selection Medium (Hybridoma Culture Medium supplemented with 0.5x HA (Sigma, cat. # A9666)), and the volume was adjusted appropriately with more Hybridoma Selection Medium, based on a final plating of 5x10° B cells total per 96-well plate and 200 μL per well. The cells were mixed gently and pipetted into 96-well plates and allowed to grow. On day 7 or 10, one-half the medium was removed, and the cells were re-fed with Hybridoma Selection Medium.
After 14 days of culture, hybridoma supernatants were screened for GPNMB specific monoclonal antibodies. In the Primary screen, the ELISA plates (Fisher, Cat. No. 12-565-136) were coated with 50 μL/well of GPNMB (1 μg/mL) in Coating Buffer (0.1 M Carbonate Buffer, pH 9.6, NaHCO3 8.4 g/L), then incubated at 40C overnight. After incubation, the plates were washed with Washing Buffer (0.05% Tween 20 in PBS) three times. 200 μL/well Blocking Buffer (0.5% BSA, 0.1% Tween 20, 0.01% Thimerosal in Ix PBS) were added and the plates were incubated at room temperature for 1 h. After incubation, the plates were washed with Washing Buffer three times. Aliquots (50 μL/well) of hybridoma supernatants and positive and negative controls were added, and the plates were incubated at room temperature for 2 h. The positive control used throughout was serum from the relevant GPNMB immunized XenoMouse® mouse and the negative control was serum from the KLH-immunized relevant strain of XenoMouse® mouse. After incubation, the plates were washed three times with Washing Buffer. 100 μL/well of detection antibody goat anti-huIgGfc-HRP (Caltag, Cat. No. Hl 0507, using concentration was 1 :2000 dilution) was added and the plates were incubated at room temperature for 1 hour. After incubation, the plates were washed three times with Washing Buffer. 100 μl/well of TMB (BioFX Lab. Cat. No. TMSK-0100-01) was added, and the plates were allowed to develop for about 10 minutes (until negative control wells barely started to show color). 50 μl/well stop solution (TMB Stop Solution (BioFX Lab. Cat. No. STPR-0100-01) was then added and the plates were read on an ELISA plate reader at a wavelength of 450 run.
The old culture supernatants from the positive hybridoma cells growth wells based on primary screen were removed completely and the IL-Ib positive hybridoma cells were suspended with fresh hybridoma culture medium and were transferred to 24-well plates. After 2 days in culture, these supernatants were ready for a secondary confirmation screen. In the secondary confirmation screen, the positives in the first screening were screened in GPNMB binding ELISA described as above, and two sets of detective system for the secondary confirmation ELISA, one set for hlgG detection, one set for human Ig kappa light chain detection (goat anti-hlg kappa-HRP, Southern Biotechnology, Cat. No. 2060-05) in order to demonstrate fully human composition for both heavy and light chains. The two sets of ELISA procedures were identical to the descriptions above except the three different detection antibodies were used separately. All positive hits from the secondary confirmation ELISA assay were counter screened for binding to immunogen. by ELISA in order to exclude those that cross-react with IL-Ia. The ELISA plates (Fisher, Cat. No. 12-565-136) were coated with 50 μL/well of irrelevant V5His-fusion protein, lug/mL in Coating Buffer (0.1 M Carbonate Buffer, pH 9.6, NaHCO3 8.4 g/L), then incubated at 4°C overnight. The remaining procedures were identical to the descriptions above. There are 33 fully human GPNMB specific monoclonal antibodies that were generated. Hybridoma supernatants were screened for binding to GPNMB by ELISA as described above in Example 2. Results are shown in Table 6. Table 6. Hybridoma anti-GPNMB activity.
Figure imgf000063_0001
Figure imgf000064_0001
Certain Hybridoma cell supernatants (29) were analyzed for binding to GPNMB by BiaCore® 2000 biosensor equipped with a research-grade CM5 sensor chip. A 1 :25 dilution of cell supernatant was passed over a protein A surface for 5 min followed by washing the surface for 10 mins. Subsequently, GPNMB was injected for 90 sec. over the surface at a concentration of 880 nM followed by dissociation. Double-referenced binding data were obtained by subtracting the signal from a control flow cell and subtracting the baseline drift of a buffer injected just prior to the antigen injection. GPNMB binding ddata for each niAb was normalized for the amount of niAb captured on each surface. Normalized, drift corrected responses were also measured. The sensorgrams were fit to a simple 1 : 1 kinetic model. The results are shown in Table 7. Sixteen of the cell supernatants contained mAb that significantly bound to GPNMB and three Mabs, 15.1, 15.2, and 15.3 showed strong binding to GPNMB. Table 7
Figure imgf000064_0002
Figure imgf000065_0001
Example 4: Binning of Antibodies
Certain antibodies, described herein were binned in accordance with the protocol described in U.S. Patent Application Publication No.20030157730. MxIiIgG conjugated beads are prepared for coupling to primary antibody. The volume of supernatant needed is calculated using the following formula: (n+10) x 50μL (where n = total number of samples on plate). Where the concentration is known, 0.5μg/mL is used. Bead stock is gently vortexed, then diluted in supernatant to a concentration of 2500 of each bead per well or 0.5X105 ImL and incubated on a shaker in the dark at RT overnight, or 2 hours if at a known concentration of 0.5μg/mL. Following aspiration, 50 μL of each bead is added to each well of filter plate, then washed once by adding 100 μL/well wash buffer and aspirating. Antigen and controls are added to filter plate 50uL/well then covered and allowed to incubate in the dark for 1 hour on shaker. Following a wash step, a secondary unknown antibody is added at 50μL/well using the same dilution (or concentration if known) as is used for the primary antibody. The plates are then incubated in the dark for 2 hours at RT on shaker followed by a wash step. Next, 50μL/well biotinylated rnxhlgG diluted 1 :500 is added and allowed to incubate in the dark for lhour on shaker at RT. Following a wash step, 50μL/well Streptavidin-PE is added at 1:1000 and allowed to incubate in the dark for 15 minutes on shaker at RT. Following a wash step, each well is resuspended in 80μL blocking buffer and read using Luminex. Results show that the monoclonal antibodies belong to distinct bins. Competitive binding by antibodies from different bins supports antibody specificity for similar or adjacent epitopes. Non competitive binding supports antibody specificity for unique epitopes. Three bins were created to further test the binding of six anti-GPNMB antibodies. Bin 1 included GPNMB antibodies (1.2.1), (1.10.1), and (2.22.1). Bin 2 included GPNMB antibodies (2.3.1) and (1.15.1), and Bin 3 included GPNMB antibody (2.10.1). The results of the binning assays are provided below in Tables 8 and 9.
Table 8
Figure imgf000066_0001
Table 9
Figure imgf000067_0001
Example 5: GPNMB Immuno histochemistry (IHC) Analysis Anti-GPNMB monoclonal antibodies were evaluated for reactivity with frozen and fixed tissue specimens. Tissue sections (5 μm) were cut from formalin fixed and paraffin embedded tissue samples and were rehydrated through incubations in xylene and a graded ethanol series terminating in PBS. Endogenous peroxidase activity was quenched in a 3% solution of hydrogen peroxide in methanol. Tissue sections were blocked in blocking buffer (5% BSA (Sigma), 1% goat serum
(Jackson Immunolabs, West Grove, PA) in PBS) for 1 hour. Primary and secondary antibodies were precomplexed in 5% BSA and 1% goat serum in PBS for 1 hour at 37° C at a molar ratio of approximately 10: 1 of anti-GPNMB or control IgG to secondary biotinylated goat anti-human IgG (Jackson Immunolabs). Complexes were blocked with a 1:2000 dilution of human serum and incubated again for 1 hour at 37° C. Tissue sections were incubated with anti-GPNMB antibody or isotype control antibody complexes diluted in blocking buffer for 1 hour. Sections were washed in 3 changes of PBS for 5 to 10 minutes each and incubated with a 1 :200 dilution of streptavidin conjugated horseradish peroxidase (Jackson Immunolabs) in blocking buffer for 30 minutes and then washed as before. Antibody was detected using DAB reagent (Vector labs). Sections were counterstained in hematoxylin (Fisher Scientific) and dehydrated through alcohol and xylene and coverslipped with permount (Fisher Scientific).
Anti-GPNMB Mabs 2.22.1 and 2.22.2 were used to stain normal and tumor human tissue microarrays (IMPATH, Los Angeles, CA). Positive staining was seen in lung, ovarian, renal, esophagus, and head & neck carcinomas, squamous cell carcinoma, melanomas and normal skin specimens. Melanoma and lung carcinomas showed the highest staining intensities with subcellular staining located in the membrane and cytoplasm. Anti-GPNMB Mab 2.10.2 also stained primary melanoma.
Anti-GPNMB antibody staining of melanoma tissue microarray showed a large proportion of melanoma cases to be positively stained as shown in Tables 10 and 11.
Table 10: anti-GPNMB Mab Melanoma Staining Intensity
Figure imgf000068_0001
On a scale of 0 (no staining) to 3 (strong staining) Table 11 : anti-GPNMB Mab Staining Frequency
Figure imgf000069_0001
* % tumor cells exhibiting positive staining
Anti-GPNMB antibody stained 10 of 14 lung squamous cell carcinoma (SCC) samples in a general oncology tissue microarray and 24 of 60 in a SCC specific array were positive.
Example 6: FACS analysis of anti-GPNMB antibody binding to Melanoma cell lines
The specificity of anti-GPNMB antibodies to cell membrane-bound GPNMB protein expressed by melanoma cancer cell line, UACC-62 was analyzed by FACS analysis. A renal cancer cell line, TKlO, which does not express GPNMB antigen was used as a negative control. Isotype matched antibody pK16.3 was used as a negative control. Cells were washed twice with PBS (Ca and Mg free), incubated with Versene at 370C until cells detached, counted and aliquoted at 1 million cells per assay tube. Cells were then washed twice and resuspended in ice-cold FACS buffer (0.01M HEPES, 0.15M NaCl, 0.1% NaN3 and 4% FBS). Primary antibody at 1 μg/mL was added to the cells. Cells were incubated on ice for 30 min, washed 2-3 times and resuspended in 1 mL of ice-cold FACS buffer. R-PE- conjugated goat anti-human antibody (Jackson ImmunoResearch Laboratory) at 1 : 100 dilution was added and cells were incubated on ice for 30 min. After washing 3 times with 1 mL of ice-cold FACS buffer, cells were fixed with 0.5-1 mL of 1 % formaldehyde in PBS and analyzed by flow cytometry.
Results expressed as Geo Mean Ratios are summarized in Table 12 and show UACC-62 cells but not TKlO cells highly express CROl 1 protein on the cell surface which was detected by 2.10.2; 2.22.1 and 1.15.1 antibodies. Table 12: Geo Mean Ratio of anti-GPNMB Staining (relative to pK16) A. Antibody B. UACC-62 Cells C. TKlO Cells
D. CROl l.2.10.2 E. 2.60 F. 1.10
G. CROl 1.2.22.1 H. 4.46 I. 1.10
J. CROl 1.1.2.2 K. 1.24 L. 0.98
M. CROI l.1.15.1 N. 7.89 O. 0.96
P. CROl 1.2.6.2 Q. 1.90 R. 1.70
To examine the relative GPNMB antigen expression among melanoma cell lines, MAb 1.15.1 antibody was used to survey a panel of 15 melanoma cell lines by FACS analysis. As shown in Table 13, 80% (12/15) of cell lines showed GPNMB antigen expression. Cell line SK-Mel-2 demonstrated the highest Geo Mean ratio among the cell lines tested. Table 13: Geo Mean Ratio of anti GPNMB Staining of Melanoma Cell Lines
Figure imgf000070_0001
Example 7: FACS analysis of anti-GPNMB MAb binding to Lymphoma and Leukemia
To determine the relative expression of GPNMB on the surface of hematopoietic malignant cells, cell lines derived from various lymphomas and leukemias were incubated with anti-GPNMB antibody and analyzed by FACS. Lymphoma or leukemia derived cells were washed twice with ice-cold FACS buffer and resuspended at 1 million cells per assay tube. MAb 1.15.1 antibody at 1 μg/mL was added to cells and cells were incubated on ice for 30 min. Cells were then washed 2-3 times and resuspended in 1 ml_ of ice-cold FACS buffer. R-PE-conjugated goat anti-human antibody at 1:100 dilution was added and cells were incubated on ice for 30 min. Cells were washed 3 times with 1 mL of ice-cold FACS buffer, fixed with 0.5-1 mL of 1 % formaldehyde in PBS and analyzed by Flow Cytometry.
Approximately half of the cell lines examined, which were derived from both myeloid and lymphoid lineages, showed GPNMB cell surface expression (Table 14). Cell line U937 demonstrated the highest Geo Mean ratio among the cell lines tested. Table 14: Geo Mean Ratio of anti-GPNMB Staining of Lymphoma and Leukemia Cells
Figure imgf000071_0001
Example 8: Detection of GPNMB protein by IP and Western Blot Analysis
Cells were washed twice with PBS (Ca and Mg free), incubated with Versene at 370C until cells detached, counted, collected and lysed in lysis buffer (0.15M NaCl, 0.02M Tris HCl, 10% glycerol, 1% NP-40, 0.0 IM EDTA and protease inhibitors containing pancreas extract, pronase, thermolysin, chymotrypsin and papain (Roche, Germany) for 30 min on ice. Supernatants were collected and protein concentrations were determined by BCA protein assay kit (Pierce, USA). Primary antibody was added to the cell lysates and incubated on ice for 3 hr followed by addition of Protein-G agarose (Amersham, USA) for 2 hr. Immunoprecipitated proteins were washed, boiled in sample buffer and resolved by A- 20% gels. For immunoblotting, proteins were transferred to PVDF membranes (Invitrogen, USA) and probed with anti-GPNMB antibody (0.5 μg/mL) followed by HRP-conjugated goat anti-human antibody (Jackson ImmunoResearch Laboratory) at 1 :4000 dilution. The immunocomplexes were detected with ECL Western blotting detection reagents (Amersham, USA).
Western blot analysis showed anti-GPNMB antibodies immunoprecipitated GPNMB protein expressed in cell lysates of UACC-62, SK-Mel5 and SK-Mel2 cell lines. The results are in concurrence with the cell surface expression determined by FACS analysis.
Example 9: anti GPNMB Antibody mediated indirect cell killing
UACC-62, a GPNMB antigen expressing cell line, and TKlO, a non-expressing cell line were plated onto flat bottom 96-well tissue culture plates (Becton Dickinson, Franklin Lakes, NJ, USA) at a density of 3000 cells per well. Once the cells reached -25% confluency, 100 ng/well of secondary antibody-toxin conjugate (goat anti -human IgG- saporin; Advanced Targeting Systems, San Diego, USA, HUM-ZAP; cat. # IT-22) was added. Anti-GPNMB MAbs 2.10.2, 2.22.1, 1.15.1 or isotype control niAb (pK16.3) were added to each well at a final concentration of 10 or 50 ng/mL. An anti-EGFR monoclonal antibody (MS-269-PABX, NeoMarkers, Fremont, CA, USA) was used as a positive primary antibody control. Chemotherapy reagent 5-FU at 600 uM was used as a positive reagent control. On day 5, the cells were trypsinized, transferred to 6-well tissue culture plates and incubated at 37 0C. Plates were examined daily and between 8-10 days, all plates were Giemsa stained and colonies were counted.
The percent viability of GPNMB positive UACC-62 after treatment is shown in Figure 2. Chemotherapy reagent 5-FU induced a complete killing whereas addition of saporin toxin-conjugated secondary antibody alone or in combination with isotype control pK16.3 antibody had no effect on cell growth for both cell lines. Both UACC-62 and TKlO cell lines express EGFR protein and addition of EGFR specific antibody at 50 ng/mL and secondary antibody toxin conjugate resulted in a complete killing of UACC-62 and TKlO cells. At the same dose, all three GPNMB specific antibodies, 2.10.2, 2.22.1 and 1.15.1 induced over 70% killing of UACC-62 cells. Anti-GPNMB antibodies 2.10.2 and 2.22.1 induced less than 5% and 1.15.1 less than 24% cell death in GPNMB negative TKlO cells. Example 10: Cell killing by Auristatin-E (AE) Conjugated anti-GPNMB Antibodies UACC-62 and TKlO cells were plated onto flat bottom 96-well tissue culture plates
(Becton Dickinson, Franklin Lakes, NJ, USA). On day 2 or cells reach -25% confluency, various concentrations (1 to 1000 ng/mL) of unconjugated and Auristatin E-conjugated antibodies (Seattle Genetics, Bothell, WA, USA), including isotype control, EGFR (NeoMarkers MS-269-PABX, Fremont, CA, USA), 2.22.1 or 2.10.2, were added to cells. MAb 2.3.1 was chosen for the isotype control in this study because it does not bind to GPNMB expressing cells as demonstrated by FACS analysis. A monoclonal antibody generated against the EGF receptor was used to demonstrate specific killing mediated by AE-conjugated antibody. On day 5, the cells were trypsinized, transferred to 6-well tissue culture plates and incubated at 370C. Plates were examined daily. On days 8-10, all plates were Giemsa stained and colonies on the plates were counted.
The percent viability in GPNMB positive UACC-62 cells and negative TKlO cells is presented in Figures 4 and 5, respectively. The results indicate that unconjugated and AE- conjugated 2.6.2 immunoconjugate had no effect on growth of both UACC-62 and TKlO cells. However, both UACC-62 and TKlO cell lines were susceptible to AE-EGFR immunoconjugate mediated cell killing in a dose-dependent fashion with over 95 % cell death at 1000 ng/mL. At the same dose, both 2.22.1 -AE and 2.10.2- AE immunoconjugates induced approximately 75 % cell death of UACC-62 cells when compared to the isotype control. The cell killing response was dose dependent. GPNMB negative TKlO cell survival was not affected by 2.22.1 -AE nor 2.10.2 -AE immunoconjugates at the same dose range. These results demonstrate the specific and cytotoxic effects of AE conjugated anti- GPNMB antibodies on antigen expressing cells. Example 11: Melanoma cells susceptible to MAbl.l5.1-AE immuno conjugate killing
Melanoma cell lines were plated onto flat bottom 96-well tissue culture plates (Becton Dickinson, Franklin Lakes, NJ, USA). On day 2 or when cells reach -25% confluency, various concentrations of unconjugated and Auristatin E-conjugated 1.15.1 were added to cells. MAb 2.6.2-AE was also used as a conjugated isotype control in this study. On day 5, the cells were trypsinized, transferred to 6-well tissue culture plates and incubated at 37 0C. Plates were examined daily. On days 8-10, all plates were Giemsa stained and colonies on the plates were counted.
The IC50 of 1.15.1 -AE mediated killing on GPNMB positive and negative cells are presented in Table 15. Unconjugated 1.15.1 and AE-conjugated 2.6.2 had no effect on growth of all the melanoma cell lines tested. However, cell lines SK-Mel2, WM-266-4, G361, UACC-257, UACC-62, RPMI-7951 and SK-Mel5 were susceptible to 1.15.1-AE mediated killing in a dose-dependent fashion. SK-Mel2 demonstrated the lowest IC50 in this study (Table 15). These results show the specific and cytotoxic effects of AE conjugated 1.15.1 on most of GPNMB expressing melanoma cells.
Table 15: Geo Mean Ratios and IC50 Values of 1.15.1-AE Killing of Melanoma Cells
Figure imgf000074_0001
Lymphoma or leukemia cell lines were mixed with methylcellulose base media (R&D Systems, USA) and in various concentrations of unconjugated and Auristatin E- coηjugated 1.15.1 antibody before plating onto 6-well tissue culture plates (Becton Dickinson, Franklin Lakes, NJ, USA). MAb 2.6.2-AE was also included as a conjugated isotype control in this study because it does not bind to GPNMB expressing cells. Plates were incubated at 37 0C and examined daily. On days 14-18, colonies on the plates were counted. The IC50 of 1.15.1-AE induced cell killing on antigen expressing cells is presented in Table 16. Unconjugated 1.15.1 and AE-conjugated 2.6.2 immunoconjugate had no effect on growth of all antigen positive hematopoietic cell lines. However, as presented in Table 16, cell lines U937, SR and THP-I derived from either myeloid or lymphoid lineage were susceptible to 1.15.1 -AE mediated killing in a dose-dependent manner with IC50 values ranging from 207 ng/mL (1.4 nJVI) to 340 ng/mL (2.4 nM). These results show the specific and cytotoxic effects of 1.15.1-AE immunoconjugate on GPNMB antigen expressing hematopoietic malignant cell lines.
Table 16: Geo Mean Ratios and IC50 Values of 1.15.1-AE Killing of Lymphoma and Leukemia Cells
Figure imgf000075_0001
Example 13: CROl l-vcMMAE Inhibits the Growth of Human SK-MEL-2 Melanoma Xenografts Leading to Complete Regression of Established Melanoma Tumo ]n Athymic Mice (Study N-386) Study N-386 was performed to assess the potency and therapeutic efficacy of the antibody-drug conjugate, CROl l-vcMMAE, against the established human SK-MEL-2 melanoma xenograft in athymic mice. Materials and Methods: Test Animals: Five- to 6-week old athymic mice (CD-I nu/nu females), used for human tumor xenografts, were obtained from Harlan Laboratories (Indianapolis, IN). Animals were housed in specific pathogen-free conditions, according to the guidelines of the Association for Assessment and Accreditation of Laboratory Animal Care International (AAALAC International). Test animals were provided pelleted food and water ad libitum and kept in a room with conditioned ventilation (HVAC), temperature (22° ± 2°C), relative humidity (55% ± 15%), and photoperiod (12 hr). All studies were carried out with approved institutional animal care and use protocols.
Human Melanoma Xenograft Models. The tumor inhibitory activity of the CROI l-MMAE immunoconjugate was measured in an anti-tumor xenograft model using athymic mice, according to published methods (see Geran et al, Cancer Chemother. Rep. 3:1-104 (1972)). Briefly, test animals were implanted subcutaneously by trocar with small fragments of a human melanoma (60-125 mg) excised from athymic mouse tumor donors. When tumors became established (10-20 days), the animals were pair-matched into groups (n= 6 mice/group), and treatment was administered by intravenous injection (tail vein). The SK-MEL-2 human melanoma (ATCC #HTB-68) was derived from a metastatic site (skin of thigh) of a 60 year old Caucasian male with malignant melanoma, and the SK- MEL-5 human melanoma (ATCC #HTB-70) was derived from a metastatic site (axillary lymph node) of a 24 year old Caucasian female with malignant melanoma (see Fogh et al, J. Natl. Cancer Inst. 59: 221-226 (1977)). Both cell lines were obtained from the American Type Culture Collection.
The effects of treatment were monitored by repetitive tumor measurements across 2 diameters with Vernier calipers; tumor size (in mg) was calculated using a standard formula, (W x L)/2, assuming a specific gravity of 1.0. Tumor size and body weights were assessed twice weekly. Mice were examined daily, however, and moribund animals were humanely euthanized if clinical indications of excessive pain or distress were noted {i.e.,
IA prostration, hunched posture, paralysis/paresis, distended abdomen, ulcerations, abscesses, seizures, and/or hemorrhages). Animals with tumors exceeding 2,000 mg were removed from the study and euthanized humanely.
Xenograft studies in the athymic mouse have been shown to effectively demonstrate anti-tumor effects for a variety of agents which have been shown subsequently to have activity against clinical cancer (Johnson et al., Br J Cancer 84: 1424-1431 (2001)). Results:
Anti-Tumor Effects In Vivo vs. SK-MEL-2 Melanoma. Based on the potency and cytotoxicity of CRO 11 -vc MMAE against GPNMB-expressing cells in vitro, the anti-tumor effects were examined in vivo.
The effects of intravenous CRO 11 -vc MMAE treatment on the growth of subcutaneous human SK-MEL-2 melanoma are shown in Figure 1. After SK-MEL-2 tumor fragments were implanted and tumors became established (day 17, 61 mg), treatment commenced with intravenous administration of: CROl l-vcMMAE (0.625 - 20 mg/kg i.v., every 4 days for a total of 4 treatments (i.e., q4d X4); saline and phosphate-buffered saline controls (i.v., q4d X4); and two known anti-tumor reference agents, vinblastine sulfate (i.v., 1.7 mg/kg, q4d X4) and paclitaxel (i.v., 24 mg/kg, q2d X4). The reference agents were administered at the maximum tolerated dose (MTD) determined in prior studies.
Tumors in animals treated with saline or PBS grew progressively until the tumor mass reached 2,000 mg at which time the animals were removed from the study and euthanized humanely. SK-MEL-2 tumors have a high "take" rate in immunocompromised hosts (97 %) and a low rate of spontaneous regression (3 %) (Dykes et al, Development of human tumor xenograft models for in vivo evaluation of new antitumor drugs, in
Immunodeficient mice in Oncology, vol. 42 (Fiebig HH and Berger DPe eds) pp 1-22, Contrib. Oncol. Basel, Karger (1992)).
Vinblastine produced a very slight, but not significant, anti-tumor effect (P < 0.20); in this and other tumor models (e.g., SK-MEL-5) vinblastine produces noticeable tumor growth inhibition, but which is only occasionally significant. Paclitaxel, however, showed significant tumor growth inhibition and tumor stasis (i.e., 100% growth inhibition) for approximately 2 weeks after treatment commenced (P < 0.0077).
The anti-tumor effects of CROl l-vcMMAE administered i.v. to SK-MEL-2-bearing mice were remarkable. At 20, 10, 5 or 2.5 mg/kg tumors rapidly diminished in size for the majority of the test animals; significant treatment effects were noted as early as 4 days after treatment commenced (P < 0.014). Tumors that regressed completely did not re-grow during the observation period (> 200 days).
The animals in this study showed no abnormal treatment effects on gross examination. Twice weekly body weight determinations showed no observable or statistically significant effects of treatment with CROl l-vcMMAE on body weight or weight gain. Conclusions:
CRO 11 -vcMMAE produces substantial, dose-dependent and reproducible anti-tumor effects that begin as tumor growth inhibition but soon lead to complete regression of established human melanoma xenografts; the regressions are long-lived and re-growth of tumors after successful therapy has not been observed.
Example 14: Sequencing of Antibodies and their Corresponding DNA
Sequences of human GPNMB mAbs-derived heavy and kappa chain transcripts from hybridomas were obtained by direct sequencing of PCR products generated from poly(A ) RNA. PCR products were also cloned into pCRII using a TA cloning kit
(Invitrogen) and both strands were sequenced using Prism dye-terminator sequencing kits and an ABI 377 sequencing instrument. Each PCR reaction used a mixture of 5' sense primers which are provided in Table 17 below. Table 17: Primers Used
Figure imgf000078_0001
All sequences were analyzed by alignments to the "V BASE sequence directory" (Tomlinson et al, MRC Centre for Protein Engineering, Cambridge, UK) using MACVECTOR® and GENEWORKS™ software programs. Example 15: Structural Analysis of Anti-GPNMB Antibodies
The variable heavy chains and the variable light chains for the antibodies shown in Table 17 were sequenced to determine their DNA and protein sequences. Antibody -1.10.2
Heavy chain variable region Nucleotide sequence
Figure imgf000079_0001
GGGGCTGGGCTGACTACTGGGGCCAGGGAACCCTGGTCACCGTCTCCTCAGCC 3 ' (SEQ ID NO : 1 )
Amino acid sequence
5'QVQLQESGPGLVKPSETLSLTCTVS GDSISNYYWS WIRQPPGKGLEWIG YFYYSGSTNYNPSLKS RVTISVDTSKNQFSLKLSSVTAADTAVYYCAR DRGWADY WGQGTLVTVSSA 3' (SEQ ID NO:2)
TABLE 18. 1.10.2 Heavy chain V region domains.
Figure imgf000079_0002
*AA Residues of SEQ ID NO:2 Light chain variable region Nucleotide sequence
5'GAAATTGTGTTGACGCAGTCTCCAGGCACCCTGTCTTTGTCTCCAGGGGAAAGGGCCACCCTCT
CCTGCAGAACCAGTCAGAGTATTAGCAGCAGCTATTTAGCCTGGTACCAGCAGAAACCTGGCCA
GGTTCCCAGGCTCCTCATCTATGGTGCTTCCAGCAGGGCCACTGGCATCCCAGACAGGTTCAGTG
GCAGTGGGTCTGGGACAGACTTCACTCTCACCATCAGCAGACTGGAGCCTGAAGATTTTGCAGTG
TATTATTGTCAGCAGTATGGTAGCTCGATCACCTTCGGCCAAGGGACACGACTGGAGATTAAACG
A 3' (SEQ ID NO: 10)
Amino acid sequence
5ΕIVLTQSPGTLSLSPGERATLSC RTSQSISSSYLA WYQQKPGQVPRLLIY GASSRAT
GIPDRFSGSGSGTDFTLTISRLEPEDFAVYYC QQYGSSIT FGQGTRLEIKR 3' (SEQ ID NO:11)
Figure imgf000079_0003
*AA Residues of SEQ ID NO: 11
Antibody -1.15.1
Heavy chain variable region
Nucleotide sequence
5'(
TCTGGTGGCTCCATCAGCAGTTTTAATTACTACTGGAGCTGGATCCGCCACCACCCAGGGAAGGGCCT
GGAGTGGATTGGGTACATCTATTACAGTGGGAGCACCTACTCCAACCCGTCCCTCAAGAGTCGAGTTACC
ATATCAGTAGACACGTCTAAGAACCAGTTCTCCCTGACGCTGAGCTCTGTGACTGCCGCGGACACGGCCG
TGTATTACTGTGCGAGAGGGTATAACTGGAACTACTTTGACTACTGGGGCCAGGGAACCCTGGTCACCGT
CTCCTCAGCC 3' (SEQ ID NO:19)
Amino acid sequence
5'QVQLQESGPGLVKPSQTLSLTCTVSGGSISSFNYYWSWIRHHPGKGLEWIGYIYYSGSTYSNPSLKSRVTIS VDTSKNQFSLTLSSVTAADTAVYYCARGYNWNYFDYWGQGTLVTVSSA 3' (SEQ ID NO:20)
TABLE 20. 1.15.1 Heavy chain V region domains.
Figure imgf000080_0001
*AA Residues of SEQ ID NO:20
Light chain variable region Nucleotide sequence
5'
GCCAGTCAGAGTGTTGACAACAACTTAGTCTGGTACCAGCAGAAACCTGGCCAGGCTCCCAGGCTCCT CATCTATGGTGCATCCACCAGGGCCACTGGTATCCCAGCCAGGTTCAGTGGCAGTGGGTCTGGGACAGAG TTCACTCTCACCATCAGTAGTCTGCAGTCTGAAGATTTTGCAGTTTATTACTGTCAGCAGTATAATAACT GGCCTCCGTGGACGTTCGGCCAAGGGACCAAGGTGGAAATCAAACGA 3' (SEQ ID NO:28)
Amino acid sequence
5'EIVMTQSPATLSVSPGERATLSCRASQSVDNNLVWYQQKPGQAPRLLIYGASTRATGIPARFSGSGSGTEFT LTISSLQSEDFAVYYCQQYNNWPPWTFGQGTKVEIKR 3' (SEQ ID NO:29)
TABLE 21. 1.15.1 Light chain V region domains.
Figure imgf000080_0002
*AA Residues of SEQ ID NO:29
Antibody -1.2.2
Heavy chain variable region
Nucleotide sequence 5' ATCACCTTGAAGGAGTCTGGTCCTACGCTGGTGAAACCCACACAGACCCTCACGCTGACC
TGCACCTTCTCTGGGTTCTCACTCAGCGCTGGTGGAGTGGGTGTGGGCTGGATCCGTCAG
CCCCCAGGAAAGGCCCTGGAGTGGCTTGCACTCATTTATTGGAATGATGATAAGCGCTAC
AGCCCATCTCTGAGGAGCAGGCTCACCATCACCAAGGACACCTCCAAAAACCAGGTGGTC
CTTACAATTACCAACATGGACCCTGTGGACACAGCCACATATTATTGTGCACACAGTCAC
TATGATTACGATTGGGGGAGTTACTTTGACTACTGGGGCCAGGGAACCCTGGTCACCGTC
TCCTCAGCC 3' (SEQ ID NO:37)
Amino acid sequence
5 ' ITLKESGPTLVKPTQTLTLTCTFS GFSLSAGGVGVG WIRQPPGKALEWLA LIYWNDDKRY SPSLRS RLTITKDTSKNQWLTITNMDPVDTATYYCAH SHYDYDWGSYFDY WGQGTLVTVSSA ( SEQ ID NO : 38 )
TABLE 22. 1.2.2 Heavy chain V region domains.
Figure imgf000081_0001
*AA Residues of SEQ ID NO:38
Light chain variable region
Nucleotide sequence
5' GATATTGTGATGACCCAGACTCCACTCTCCCTGCCCGTCACCCCTGGAGAGCCGGCCTCC
ATCTCCTGCAGGTCTAGTCAGAGCCTCTTGGATAGTGATGATGGAAACACCTATTTGGAC
TGGTACCTGCAGAAGCCAGGACAGTCTCCACAGCTCCTGATCTATACGCTTTCCTATCGG
GCCTCTGGAGTCCCAGACAGGTTCAGTGGCAGTGGGTCAGGCACTGATTTCACACTGAAC
ATCAGCAGGGTGGAGGCTGAGGATGTTGGAGTTTATTACTGCATGCAACGTATAGAGTTT
CCTATCACCTTCGGCCAAGGGACACGACTGGAGATTAAACGA 3' (SEQ ID NO:46)
Amino acid sequence
5' DIVMTQTPLSLPVTPGEPASISC RSSQSLLDSDDGNTYLD WYLQKPGQSPQLLIY TLSYRAS GVPDRFSGSGSGTDFTLNISRVEAEDVGVYYC MQRIEFPIT FGQGTRLEIKR 3' (SEQ ID NO:47) TABLE 23. 1.2.2 Light chain V region domains.
Figure imgf000081_0002
*AA Residues of SEQ ID NO:47
Antibody -1.7.1
Heavy chain variable region
Nucleotide sequence
5' CAGGTGCAGCTGCAGGAGTCGGGCCCAGGACTGGTGAAGCCTTCACAGACCCTGTCCCTC ACCTGCACTGTCTCTGGTGGCTCCATCAGCAGTGCTAATTACTACTGGACCTGGATCCGC CAGCACCCAGGGAAGGGCCTGGAGTGGATTGGGTACATCTATTACAGTGGGAGCACCTAC TGCAACCCGTCCCTCAAGAGTCGAGTTATCATATCAGTAGACACGTCTAAGAACCAGTTC TCCCTGAAGCTGAGCTCTGTGACTGCCGCGGACACGGCCGTGTATTACTGTGCGAGAGGG TATAACTGGAACTACTTTGACTACTGGGGCCAGGGAACCCTGGTCACCGTCTCCTCAGCC 3' (SEQ ID NO:55)
Amino acid sequence
5' QVQLQESGPGLVKPSQTLSLTCTVS GGSISSANYYWT WIRQHPGKGLEWIG YIYYSGSTY CNPSLKS RVIISVDTSKNQFSLKLSSVTAADTAVYYCAR GYNWNYFDY WGQGTLVTVSSA 3' (SEQ ID NO:56)
Figure imgf000082_0001
*AA Residues of SEQ ID NO:56 Light chain variable region Nucleotide sequence
5'( GCi
TGGTGCATCCACCAGGGCCACTGGTATCCCAGCCAGGTTCAGTGGCAGTGGGTCTGGGACAGAGTTCACTCTCA CCATCAGCAGCCTGCAGTCTGAAGATTTTGCAGTTTATTACTGTCAGCAGTATAATAAGTGGCCTCCGTGGACG TTCGGCCAAGGGACCAAGGTGGAAATCGAACGAACT 3' (SEQ ID NO:64)
Amino acid sequence
5 ' DIVMTQSPATLSVSPGERATLSC RASQSVSSNLA WYQERPGQAPRLLIY GASTRAT GIPARFSGSGSGTEFTLTISSLQSEDFAVYYC QQYNKWPPWT FGQGTKVEIER 3 ' ( SEQ ID NO : 65 )
Figure imgf000082_0002
*AA Residues of SEQ ID NO:65
Antibody -2.10.2
Heavy chain variable region
Nucleotide sequence
5' CAGCTGGTGGAGTCTGGGGGAGGCGTGGTCCAGCCTGGGAGGTCCCTGAGACTCTCCTGT
GCAGCCTCTGGATTCGCCTTCAGTAGCTATGGCATGCACTGGGTCCGCCAGGCTCCAGGC
AAGGGGCTGGAGTGGGTGGCAGTTATATCATATGATGGAAATAATAAATACTATGCAGAC
TCCGTGAAGGGCCGATTCACCATCTCCAGAGACAATTCCAAGAACACGCTGTATCTGCAA
ATGAACAGCCTGAGAGCTGAGGACACGGCTGTGTATTACTGTGCGAGAGATCTAGTGGTT CGGGGAATTAGGGGGTACTACTACTACTTCGGTATGGACGTCTGGGGCCAAGGGACCACG GTCACCGTCTCCTCAGCC 3' (SEQ ID NO:73)
Amino acid sequence
5 ' QLVESGGGWQPGRSLRLSCAAS GFAFSSYGMH WVRQAPGKGLEWVA VISYDGNNKYYAD SVKG RFTISRDNSKNTLYLQMNSLRAEDTAVYYCAR DLWRGIRGYYYYFGMDV WGQGTT VTVSSA 3 ' ( SEQ ID NO : 74 )
TABLE 26. 2.10.2 Heavy chain V region domains
Figure imgf000083_0001
* AA Residues of SEQ ID NO:74
Light chain variable region Nucleotide sequence
5' GATATTGTGATGACTCAGTCTCCACTCTCCCTGCCCGTCACCCCTGGAGAGCCGGCCTCC
ATCTCCTGCAGGTCTAGTCAGAGCCTCCTGCATAGTAATGGATACAACTATTTGGATTGG
TACCTGCAGAAGCCAGGGCAGTCTCCACAGCTCCTGATCTATTTGGGTTCTAATCGGGCC
TCCGGGGTCCCTGACAGGTTCAGTGGCAGTGGATCAGGCACAGATTTTACACTGAAAATC
AGCAGAGTGGAGGCTGAGGATGTTGGGGTTTATTACTGCATGCAAGGTCTACAAACTCCG
ATCACCTTCGGCCAAGGGACACGACTGGAGATTAAACGA 3' (SEQ ID NO:82)
Amino acid sequence
5 ' DIVMTQSPLSLPVTPGEPASISC RSSQSLLHSNGYNYLD WYLQKPGQSPQLLIY LGSNRAS GVPDRFSGSGSGTDFTLKISRVEAEDVGVYYC MQGLQTPIT FGQGTRLEIKR 3 ' (SEQ ID NO : 83 ) TABLE 27. 2.10.2 Light chain V region domains.
Figure imgf000083_0002
*AA Residues of SEQ ID NO:83
Antibody - 2.15.1
Heavy chain variable region
Nucleotide sequence
5' CAGGTGCAGCTGGTGGAGTCTGGGGGAGGCGTGGTCCAGCCTGGGAGGTCCCTGAGACTC
TCCTGTGCAGCGTCTGGATTCACCTTCAGTAACTATGGCATTCACTGGGTCCGCCAGGCT
CCAGGCAAGGGGCTGGAGTGGGTGGCAGTTATATGGTTTGATGGACGTAATAAATACTAT
GCAGACTCCGTGAAGGGCCGATTCACCATCTCCAGAGACAATTCCAAGAACACGCTGTAT
CTGCAAATGAACAGCCTGAGAGCCGAGGACGCGGCTGTGTATTACTGTGCGAGAGATCCC
TTTGACTATGGTGACTCCTTCTTTGACTACTGGGGCCAGGGCACCCTGGTCACCGTCTCC
TCAGCC 3' ( (SSEEQQ IIDD NNOO::9911)) Amino acid sequence
5' QVQLVESGGGWQPGRSLRLSCAAS GFTFSNYGIH WVRQAPGKGLEWVA VIWFDGRNKYY ADSVKG RFTISRDNSKNTLYLQMNSLRAEDAAVYYCAR DPFDYGDSFFDY WGQGTLVTVSSA 3' (SEQ ID NO:92)
TABLE 28. 2.15.1 Heav chain V re ion domains
Figure imgf000084_0001
*AA Residues of SEQ ID NO: 92 Light chain variable region Nucleotide sequence
55' CTGACTCAGTCTCCATCCTCCCTGTCTGCATCTGTAAGAGACAGAGTCACCATCACTTGC CGGGCGAGTCAGGACATTAGCAATTATTTAGCCTGGTATCAGCAGAAACCAGGGAAAGTT CCTAATCTCCTGATCTATGCTGCATCCACTTTGCAATCAGGGGTCCCATCTCGGTTCAGT GGCAGTGGATCTGGGACAGATTTCACTCTCACCATCAGCAGCCTGCAGCCTGAAGATGTT GCAACTTATTACTGTCAAAAGTATAACAGTGCCCCGCTCACTTTCGGCGGAGGGACCAAG GTGGAGATCAAACGA 3' (SEQ ID NO:100)
Amino acid sequence
5 ' LTQSPSSLSASVRDRVTITC RASQDISNYLA WYQQKPGKVPNLLIY AASTLQS GVPSRFS GSGSGTDFTLTISSLQPEDVATYYC QKYNSAPLT FGGGTKVEIKR 3 ' ( SEQ ID NO : 101 )
TABLE 29. 2.15.1 Light chain V region domains.
Figure imgf000084_0002
*AA Residues of SEQ ID NO: 101
Antibody-2.16.1
Heavychainvariableregion
Nucleotidesequence
5' CAGGTGCAGCTGGTGGAGTCTGGGGGAGGCTTGGTCAAGCCTGGAGGGTCCCTGAGACTC
TCCTGTGCAGCCTCTGGATTCACCTTCAGTGACTACTACATGACCTGGATCCGCCAGGCT
CCAGGGAAGGGGCTGGAGTGGGTTTCATACATTAGTATTAGTGGTAGTATCACACACTAC
GCAGACTCAGTGAΆGGGCCGATTCACCATGTCCAGGGACAACGCCAAGAACTCACTGTAT
CTGCAAATGAACAGCCTGAGAGCCGAGGACACGGCCGTGTATTACTGTGCGAGAGACGGA
GCAGCAGCTGGTACGGATGCTTTTGATATCTGGGGCCACGGGACAAAGGTCACCGTCTCT TCAGCC 3 ' ( SEQ ID NO : 109 )
Amino acid sequence
5 ' QVQLVESGGGLVKPGGSLRLSCAAS GFTFSDYYMT WIRQAPGKGLEWVS YISI SGSITHY ADSVKG RFTMSRDNAKNSLYLQMNSLRAEDTAVYYCAR DGAAAGTDAFDI WGHGTKVTVSSA ( SEQ ID NO : 110 )
TABLE 30. 2.16.1 Heavy chain V region domains.
Figure imgf000085_0002
*AA Residues of SEQ ID NO: 1 10
Light chain variable region Nucleotide sequence
Figure imgf000085_0001
Amino acid sequence 5'EIVMTQSPATLSVSPGDRATLSC RASQNVSSNLA WYQQKPGQAPRLLIF GASTRAT
GIPARFSGSGSGTEFTLTISSLQSEDFAVYYC QQYHYWPT FGPGTKVDIKR 3' (SEQ ID NO:119)
TABLE 31. 2.16.1 Li ht chain V region domains.
Figure imgf000085_0003
*AA Residues of SEQ ID NO: 119
Antibody -2.17.1
Heavy chain variable region
Nucleotide sequence
5' CAGCTGGTGCAGTCTGGGGCTGAGGTGAAGAAGCCTGGGGCCTCAGTGAAGGTCTCCTGC
AAGGCTTCTGGATACACCTTCACCGGCTTCTATATGCACTGGGTGCGACAGACCCCTGGA
CAAGGGCTTGAGTGGATGGGATGGATCAACCCTAACAGTGGTGGCACATATTATGTACAG
AAGTTTCAGGGCAGGGTCACCATGACCAGGGACACGTCCATCAGCACAGTCTACATGGAG
CTGAGCAGGTTGAGATCTGACGACACGGCCGTATATTACTGTGCGAGAGATGGGTATAGC
AGTGGAGAGGACTGGTTCGACCCCTGGGGCCAGGGAACCCTGGTCACCGTCTCCTCAGCC 3' (SEQ ID
NO:127)
Amino acid sequence
5' QLVQSGAEVKKPGASVKVSCKAS GYTFTGFYMH WVRQTPGQGLEWMG WINPNSGGTYYVQ KFQG RVTMTRDTSISTVYMELSRLRSDDTAVYYCAR DGYSSGEDWFDP WGQGTLVTVSSA 3' (SEQ ID NO:128) TABLE 32. 2.17.1 Heavy chain V region domains.
Figure imgf000086_0002
*AA Residues of SEQ ID NO: 128
Light chain variable region Nucleotide sequence
Figure imgf000086_0001
CTTCCGCTCACTTTCGGCGGAGGGACCAAGGTGGAGATCAAACGA 3 ' ( SEQ ID NO : 136 )
Amino acid sequence
5'DIVMTQTPLSLSVTPGQPASISC KSSQSLLHSGGKTYLY WYLQRPGQPPQLLIY EVSNRFS GVPDRFSGSGSGTDFTLKISRVEAEDVGVYYC MQSIHLPLT FGGGTKVEIKR 3' (SEQ ID NO:137)
Figure imgf000086_0003
*AA Residues of SEQ ID NO: 137
Antibody -2.21.1
Heavy chain variable region
Nucleotide sequence
5' CAGGTGCAGCTGGAGCAGTCGGGGGGAGGCCTGGTCAAGCCTGGGGGGTCCCTGAGATTC
TCCTGTGCAGCCTCTGGATTCACCTTCAGTAGCTATAGCATGAACTGGGTCCGCCAGGCT
CCAGGGAAGGGGCTGGAGTGGGTCTCATTCATTAGTAGTAGTAGTAGTTACATATACTAC
GCAGACTCAGTGAAGGGCCGATTCACCATCTCCAGAGACAACGCCAAGAACTCACTGTAT
CTGCAAATGAACAGCCTGAGAGCCGAGGACACGGCTGTGTATTACTGTGCGAGAGAGGAC
TGGGTGGGAGCTACCTTTGACTACTGGGGCCAGGGAACCCTGGTCACCGTCTCCTCAGCC 3' (SEQ ID
NO:145)
Amino acid sequence
5' QVQLEQSGGGLVKPGGSLRFSCAAS GFTFSSYSMN WVRQAPGKGLEWVS FISSSSSYIYY ADSVKG RFTISRDNAKNSLYLQMNSLRAEDTAVYYCAR EDWVGATFDY WGQGTLVTVSSA 3' (SEQ ID NO:146)
Figure imgf000087_0001
*AA Residues of SEQ ID NO: 146
Light chain variable region Nucleotide sequence
5' GACATTCAGCTGACCCAGTCTCCATCCTCCCTGTCTGCATCTGTAGGAGACAGAGTCACC
ATCACTTGTCGGGCGAGTCAGGGCATTAGGAATTATTTAGCCTGGTATCAGCAGAAACCA
GGGAAAGTTCCTAAGCTCCTGATCTATGCTGCTTCCGCTTTGAAATTAGGGGTCCCATCT
CGGTTCAGTGGCAGTGGATCTGGGACAGATTTCACTCTCACCATCAGCAGCCTGCAGCCT
GAAGATGTTGCAACTTATTACTGTCAAAAGTATAACAGTGCCCCGATCACCTTCGGCCAA
GGGACACGACTGGACATTAAACGA 3' (SEQ ID NO:154)
Amino acid sequence
5 ' DIQLTQSPSSLSASVGDRVTITC RASQGIRNYLA WYQQKPGKVPKLLIY AASALKL GVPS RFSGSGSGTDFTLTISSLQPEDVATYYC QKYNSAPIT FGQGTRLDIKR 3 ' ( SEQ ID NO : 155 )
TABLE 35. 2.21.1 Light chain V region domains.
Figure imgf000087_0002
*AA Residues of SEQ ID NO: 155
Antibody -2.22.1
Heavy chain variable region
Nucleotide sequence
5' CAGGTGCAGCTGGAGCAGTCGGGCCCAGGACTGGTGAAGCCTTCACAGAACCTGTCCCTC
ACCTGCACTGTCTCTGGTGGCTCCATCAGCAGTGGTGGTTATTTCTGGAGCTGGATCCGC
CAGCACCCAGGGAAGGGCCTGGAGTGGATTGGGTACATCTATTACAGTGGGAACACCTAC
TACAACCCGTCCCTCAAGAGTCGAGTTACCATATCAGTTGACACGTCTAAGAACCAGTTC
TCCCTGAAACTGAGCTCTGTGACTGCCGCGGACACGGCCGTGTATTACTGTGCGAGAGAC
TATTACTATGATACTAGTGGTTTTTCCTACCGTTACGACTGGTACTACGGTATGGACGTC
TGGGGCCAAGGGACCACGGTCACCGTCTCCTCAGCC 3' (SEQ ID NO:163)
Amino acid sequence
5 ' QVQLEQSGPGLVKPSQNLSLTCTVS GGSISSGGYFWS WIRQHPGKGLEWIG YIYYSGNTY YNPSLKS RVTISVDTSKNQFSLKLSSVTAADTAVYYCAR DYYYDTSGFSYRYDWYYGMDV WGQGTTVTVSSA 3 ' ( SEQ ID NO : 164 ) TABLE 36. 2.22.1 Heavy chain V region domains.
Figure imgf000088_0001
*AA Residues of SEQ ID NO: 164
Light chain variable region Nucleotide sequence
5' GACATCCAGCTGACCCAGTCTCCATCCTCCCTGTCTGCATCTGTAGGAGACAGAGTCACC
ATCACTTGCCGGGCAAGTCAGGGCATTAGAAATGATTTAGGCTGGTATCAGCAGAAACCA
GGGAAAGCCCCTAAGCGCCTGATCTATGCTGCATCCAGTTTGCAAAATGGGGTCCCATCA
AGGTTCAGCGGCAGTGGATCTGGGACAGAATTCACTCTCACAATCAGCAGCCTGCAGCCT
GAAGATTTTGCAACTTATTACTGTCTACAACATAATACTTACCCGGCGTTCGGCCAAGGG
ACCAAGGTGGAAATCAAACGA 3' (SEQ ID NO: 172)
Amino acid sequence
5' DIQLTQSPSSLSASVGDRVTITC RASQGIRNDLG WYQQKPGKAPKRLIY AASSLQN GVPS RFSGSGSGTEFTLTISSLQPEDFATYYC LQHNTYPA FGQGTKVEIKR 3' (SEQ ID NO:173)
Figure imgf000088_0002
*AA Residues of SEQ ID NO: 173
Antibody - 2.24.1
Heavy chain variable region
Nucleotide sequence
5' CAGCTGGTGCAGTCTGGAGCAGAAGTGAAAAAGCCCGGGGAGTCTCTGAAGATCTCCTGT
CAGGGTTCTGGATACATCTTTACCAACTACTGGATCGGCTGGGTGCGCCAGATGCCCGGG
AAAGGCCTGGAGTGGATGGGGGTCATCTATCCTGATGACTCTGATACCAGATACAGCCCG
TCCTTCCAAGGCCAGGTCACCATCTCAGCCGACAAGTCCATCAGCACCGCCTACCTGCAG
TGGAGCAGCCTGAAGGCCTCGGACACCGCCATATATTACTGTGCGAGACAAAAATGGCTA
CAACACCCCTTTGACTACTGGGGCCAGGGAACCCTGGTCACCGTCTCCTCAGCC 3' (SEQ ID NO:181)
Amino acid sequence
5 ' QLVQSGAEVKKPGESLKISCQGS GYIFTNYWIG WVRQMPGKGLEWMG VIYPDDSDTRYSP
SFQG QVTISADKSI STAYLQWSSLKASDTAIYYCAR QKWLQHPFDY WGQGTLVTVSSA 3 ' ( SEQ ID
NO : 182)
Figure imgf000089_0001
*AA Residues of SEQ ID NO: 182
Light chain variable region Nucleotide sequence
5' GAAATTGTGTTGACGCAGTCACCAGGCACCCTGTCTTTGTCTCCAGGGGAAAGAGTCACC
CTCTCATGCAGGGCCAGTCAGAGTGTTAGCAGCAGATACTTAGCCTGGTACCAGCAGAAA
CCTGGCCAGGCTCCCAGGCTCCTCATCTATGGTGCATCCAGCAGGGCCACTGGCATCCCA
GACAGGTTCAGTGGCAGTGGGTCTGGGACAGACTTCACTCTCACCATCAGCAGACTGGAG
CCTGAAGATTTTGCAGTTTATTACTGTCAGCAGTATGGTAGCTCACCTCGGACGTTCGGC
CAAGGGACCAAGGTGGAAATCAAACGA 3' (SEQ ID NO:190)
Amino acid sequence
5 ' EIVLTQSPGTLSLSPGERVTLSC RASQSVSSRYLA WYQQKPGQAPRLLIY GASSRAT GIP DRFSGSGSGTDFTLTISRLEPEDFAVYYC QQYGSSPRT FGQGTKVEIKR 3 ' ( SEQ ID NO : 191 )
Figure imgf000089_0002
*AA Residues of SEQ ID NO: 191
Antibody -2.3.1
Heavy chain variable region
Nucleotide sequence
5'CAGGTGCAGCTGGTGCAGTCTGGGGCTGAGGTGAAGAAGCCTGGGGCCTCAGTGAAGGTCTCC
TGCAAGGCTTCTGGATACACCTTCACCGGCTACTATATGCACTGGGTGCGACAGGCCCCTGGACA
AGGGCTTGAGTGGATGGGATGGATCAACCCTAACAGTGGTGGCACAAACTATGCACAGAAGTTT
CAGGACAGGGTCACCATGACCAGGGACACGTCCATCAGCACAGCCTACATGGAGCTGAGCAGGC
TGAGATCTGACGACACGGCCGTGTATTACTGTGCGAGAGATTTCTTTGGTTCGGGGAGTCTCCTC
TACTTTGACTACTGGGGCCAGGGAACCCTGGTCACCGTCTCCTCAGCC 3' (SEQ ID NO:199)
Amino acid sequence
5'QVQLVQSGAEVKKPGASVKVSCKASGYTFTGYYMHWVRQAPGQGLEWMGWINPNSGGTNYAQKFQD RVTMTRDTSISTAYMELSRLRSDDTAVYYCARDFFGSGSLLYFDYWGQGTLVTVSSA 3' (SEQ ID NO:200) TABLE 40. 2.3.1 Heavy chain V region domains.
Figure imgf000090_0002
*AA Residues of SEQ ID NO:200
Light chain variable region Nucleotide sequence
Figure imgf000090_0001
C CTTTTCCCCGGCCTTCCAACCTTTTTTCCGGGGCCGGGGAAGGGGGGAACCCCAAAAGGGGTTGGGGAAGGAATTCCAAAAAACCGGAA 3' (SEQ ID NO:208)
Amino acid sequence
5'DIVMTQTPLSLSVTPGQPASISC KSSQSLLHSGGKTYLY WYLQRPGQPPQLLIY EVSNRFS GVPDRFSGSGSGTDFTLKISRVEAEDVGVYYC MQSIHLPLT FGGGTKVEIKR 3' (SEQ ID NO:209) TABLE 41. 2.3.1 Light chain V region domains.
Figure imgf000090_0003
*AA Residues of SEQ ID NO:209
Antibody -2.6.1
Heavy chain variable region
Nucleotide sequence
5'CAGGTGCAGCTGGTGCAGTCTGGGGCTGAGGTGAAGAAGCCTGGGGCCTCAGTGAAGGTCTCC
TGCAAGGCTTCTGGATACACCTTCACCGGCTACTATATGCACTGGGTGCGACAGGCCCCTGGACA
AGGGCTTGAGTGGATGGGATGGATCAACCCTAACAGTGGTGGCACAAACTATGCACAGAAGTTT
CAGGACAGGGTCACCATGACCAGGGACACGTCCATCAGCACAGCCTACATGGAGCTGAGCAGGC
TGAGATCTGACGACACGGCCGTGTATTACTGTGCGAGAGATTTCTTTGGTTCGGGGAGTCTCCTC
TACTTTGACTACTGGGGCCAGGGAACCCTGGTCACCGTCTCCTCAGCC 3' (SEQ ID NO:309)
Amino acid sequence
5'QVQLVQSGAEVKKPGASVKVSCKAS GYTFTGYYMH WVRQAPGQGLEWMG WINPNSGGTNYAQKFQD RVTMTRDTSISTAYMELSRLRSDDTAVYYCAR DFFGSGSLLYFDY WGQGTLVTVSSA 3' (SEQ ID NO:310)
Figure imgf000091_0001
*AA Residues of SEQ ID NO:310
Light chain variable region Nucleotide sequence
5' TCTAGTCAGAGCCTCCTGCATAGTGGTGGAAAGACCTATTTGTATTGGTACCTGCAGAGGCCAGGCCAGCCTCC
CTTCCGCTCACTTTCGGCGGAGGGACCAAGGTGGAGATCAAACGA 3' (SEQ ID NO:318)
Amino acid sequence
5'DIVMTQTPLSLSVTPGQPASISC KSSQSLLHSGGKTYLY WYLQRPGQPPQLLIY EVSNRFS GVPDRFSGSGSGTDFTLKISRVEAEDVGVYYC MQSIHLPLT FGGGTKVEIKR 3' (SEQ ID NO:319) TABLE 43. 2.6.1 Li ht chain V region domains.
Figure imgf000091_0002
*AA Residues of SEQ ID NO:319
Antibody-2.7.1
Heavychainvariableregion
Nucleotidesequence
5' CAGGTGCAGCTGGAGCAGTCGGGGGGAGGCGTGGTCCAGCCTGGGAGGTCCCTGAGACTC
TCCTGTGCAGCGTCTGGATTCACCTTCAATAACTATGGCATGCACTGGGTCCGCCAGGCT
CCAGGCAAGGGGCTGGAGTGGGTGGCAGTTATATGGTATGATGGAAGTAATAAATACTAT
GCAGACTCCGTGAAGGGCCGATTCACCATCTCCAGAGACAATTCCAAGAACACGCTGTAT
CTGCAAΆTGAACAGCCTGAGAGCCGAGGACACGGCTGTGTATTACTGTGCGAAAGATGAG
GAATACTACTATGTTTCGGGGCTTGACTACTGGGGCCAGGGAACCCTGGTCACCGTCTCC
TCAGCC 3' (SEQ ID NO:217)
Amino acid sequence
5 ' QVQLEQSGGGWQPGRSLRLSCAAS GFTFNNYGMH WVRQAPGKGLEWVA VIWYDGSNKYY ADSVKG RFTISRDNSKNTLYLQMNSLRAEDTAVYYCAK DEEYYYVSGLDY WGQGTLVTVSSA 3 ' ( SEQ ID NO : 218 ) TABLE 44. 2.7.1 Heav chain V re ion domains.
Figure imgf000092_0001
*AA Residues of SEQ ID NO:218
Light chain variable region Nucleotide sequence
5' CTGACTCAGTCTCCATCCTCCCTGTCTGCATCTGTAAGAGACAGAGTCACCATCACTTGC
CGGGCGAGTCAGGACATTAGCAATTATTTAGCCTGGTATCAGCAGAAACCAGGGAAAGTT
CCTAATCTCCTGATCTATGCTGCATCCACTTTGCAATCAGGGGTCCCATCTCGGTTCAGT
GGCAGTGGATCTGGGACAGATTTCACTCTCACCATCAGCAGCCTGCAGCCTGAAGATGTT
GCAACTTATTACTGTCAAAAGTATAACAGTGCCCCGCTCACTTTCGGCGGAGGGACCAAG
GTGGAGATCAAACGA 3' (SEQ ID NO:226)
Amino acid sequence
5 ' LTQSPSSLSASVRDRVTITC RASQDISNYLA WYQQKPGKVPNLLIY AASTLQS GVPSRFS GSGSGTDFTLTISSLQPEDVATYYC QKYNSAPLT FGGGTKVEIKR 3 ' ( SEQ ID NO : 227 )
Figure imgf000092_0002
*AA Residues of SEQ ID NO:227 Antibody - 2.8.1
Heavy chain variable region Nucleotide sequence
5' CAGATCACCTTGAAGGAGTCTGGTCCTACGCTGGTGACACCCACACAGACCCTCACGCTG ACCTGCACCTTCTCTGGGTTCTCACTCAGCACTGGTGGAATGGGTGTGGGCTGGATCCGT CAGCCCCCAGGAAAGGCCCTGGACTGGCTTACACTCATTTATTGGAATGATGATAAGCAC TACAGCCCATCTCTGAAGAGCAGGCTTACCATCACCAAGGACACCTCCAAAAACCAGGTG GTCCTTAGAATGACCAACATGGACCCTGTGGACACAGCCACTTATTACTGTGCACACCTG CATTACGATATTTTGACTGGTTTTAACTTTGACTACTGGGGCCAGGGAACCCTGGTCACC GTCTCCTCAGCC 3' (SEQ ID NO:235) Amino acid sequence
5 ' QITLKESGPTLVTPTQTLTLTCTFS GFSLSTGGMGVG WIRQPPGKALDWLT LIYWNDDKH YSPSLKS RLTITKDTSKNQWLRMTNMDPVDTATYYCAH LHYDILTGFNFDY WGQGTLVTVSSA 3 ' ( SEQ ID NO : 236 ) TABLE 46. 2.8.1 Heav chain V re ion domains.
Figure imgf000093_0002
*AA Residues of SEQ ID NO:236 Light chain variable region Nucleotide sequence
Figure imgf000093_0001
GAGTTTCCGCTCACTTTCGGCGGAGGGACCAAGGTGGAGATCAAACGA 3 ' ( SEQ ID NO : 244 )
Amino acid sequence
5'DIVMTQTPLSLPVTPGEPASISC RSSQSLLDSDDGNTYLD WYLQKPGQSPQLLIY TLSYRAS GVPDRFSGSGSGTDFTLKISRVEAEDVGVYYC MQRIEFPLT FGGGTKVEIKR 3' (SEQ ID NO:245)
TABLE 47. 2.8.1 Li ht chain V re ion domains.
Figure imgf000093_0003
*AA Residues of SEQ ID NO:245
Example 16:Use of anti-GPNMB Antibodies as a Diagnostic Agent Detection of GPNMB antigen in a sample:
The following is a protocol for an Enzyme-Linked Immunosorbent Assay (ELISA) for the detection of GPNMB antigen in a sample. In the assay, wells of a microtiter plate, such as a 96-well microtiter plate or a 384-well microtiter plate, are adsorbed for several hours with a first fully human monoclonal antibody directed against GPNMB. The immobilized antibody serves as a capture antibody for any of the GPNMB that may be present in a test sample. The wells are rinsed and treated with a blocking agent such as milk protein or albumin to prevent nonspecific adsorption of the analyte.
Subsequently the wells are treated with a test sample suspected of containing the GPNMB antigen, or with a solution containing a standard amount of GPNMB antigen. Such a sample may be, for example, a serum sample from a subject suspected of having levels of circulating GPNMB considered to be diagnostic of a pathology.
After rinsing away the test sample or standard, the wells are treated with a second fully human monoclonal anti-GPNMB antibody that is labeled by conjugation with biotin. The labeled anti-GPNMB antibody serves as a detecting antibody. After rinsing away excess second antibody, the wells are treated with avidin-conjugated horseradish peroxidase (HRP) and a suitable chromogenic substrate. The concentration of the antigen in the test samples is determined by comparison with a standard curve developed from the standard samples. This ELISA assay provides a highly specific and very sensitive assay for the detection of the GPNMB antigen in a test sample. Determination of GPNMB antigen concentration in patients:
A sandwich ELISA can also be used to quantify GPNMB levels in human serum. The 2 fully human monoclonal anti-GPNMB antibodies used in the sandwich ELISA, recognize different epitopes on the GPNMB molecule. The ELISA is performed as follows: 50 μl of capture anti-GPNMB antibody in coating buffer (0.1 M NaHCθ3, pH 9.6) at a concentration of 2 μg/mL is coated on ELISA plates (Fisher). After incubation at 4°C overnight, the plates are treated with 200 μl of blocking buffer (0.5% BSA, 0.1% Tween 20, 0.01% Thimerosal in PBS) for 1 hr at 25°C. The plates are washed (3x) using 0.05% Tween 20 in PBS (washing buffer, WB). Normal or patient sera (Clinomics, Bioreclaimation) are diluted in blocking buffer containing 50% human serum. The plates are incubated with serum samples overnight at 4°C, washed with WB, and then incubated with 100 μl/well of biotinylated detection anti-GPNMB antibody for 1 hr at 250C. After washing, the plates are incubated with HRP-Streptavidin for 15 min, washed as before, and then treated with 100 μl/well of o-phenylenediamine in H2O2 (Sigma developing solution) for color generation. The reaction is stopped with 50 μl/well of H2SO4 (2M) and analyzed using an ELISA plate reader at 492 ran. Concentration of GPNMB antigen in serum samples is calculated by comparison to dilutions of purified GPNMB antigen using a four parameter curve fitting program. Staging of cancer in a patient:
It will be appreciated that based on the results set forth and discussed in the above diagnostic examples, it is possible to stage a cancer in a subject based on expression levels of the GPNMB antigen. For a given type of cancer (e.g., melanoma), samples of blood are taken from subjects diagnosed as being at various stages in the progression of the disease, and/or at various points in the therapeutic treatment of the cancer. The concentration of the GPNMB antigen present in the blood samples is determined using a method that specifically determines the amount of the antigen that is present. Such a method includes an ELISA method, such as the method described in the previous diagnostic examples. Using a population of samples that provides statistically significant results for each stage of progression or therapy, a range of concentrations of the antigen that may be considered characteristic of each stage is designated. In order to stage the progression of the cancer in a subject under study, or to characterize the response of the subject to a course of therapy, a sample of blood is taken from the subject and the concentration of the GPNMB antigen present in the sample is determined. The concentration so obtained is used to identify in which range of concentrations the value falls. The range so identified correlates with a stage of progression or a stage of therapy identified in the various populations of diagnosed subjects, thereby providing a stage in the subject under study. Example 17: Diagnosing Cancer With Antibodies Against GPNMB
A subject suspected of having an ovarian cancer tumor is identified and a tissue sample from the suspected tumor is removed for testing. The removed tissue is then contacted with anti-GPNMB antibodies having a colorimetric label. A determination is made of whether the anti-GPNMB antibodies bind specifically to the removed tissue. Binding is indicative of cancereous tissue while the absense of binding is indicative of noncancerous tissue. The patient's conditition is diagnosed accordingly to facilitate subsequent testing, counseling, and/or treatment. Example 18: Treating Cancer With Antibodies Against GPNMB
Targeting GPNMB on tumor cells is useful to treat a subject at risk for or afflicted with cancer. Such a subject would benefit from treatment with an anti-GPNMB antibody of the present invention. Typically, antibodies are administered in an outpatient setting by weekly administration at about 0.1-1.0 mg/kg dose by slow intravenous (IV) infusion. The appropriate therapeutically effective dose of an antibody is selected by a treating clinician and would range approximately from 1 iig/kg to 20mg /kg, from 1 μ_g/kg to 10 mg/kg, from 1 μ.g/kg to 1 mg/kg, from 10 μg/kg to 1 mg/kg, from 10 j±g/kg to 100 μg/kg, from 100 jxg/kg to 1 mg/kg, and from 500 j±g/kg to 5 mg/kg. The antibodies are also used to prevent and/or to reduce severity and/or symptoms of disease associated with GPNMB-related disorders.
To test the clinical efficacy of antibodies in humans, individuals with cancer, particularly, but not limited to ovarian, lung or colon carcinoma are identified and randomized into treatment groups. Treatment groups include a group not receiving antibody treatment and groups treated with different doses of anti-GPNMB antibody. Individuals are followed prospectively and individuals receiving antibody treatment exhibit an improvement in their condition.
Example 19: The Specificity of the Anti-tumor Effects of CROll-vcMMAE ( CROIl- ONC-I)
The study was performed to determine the anti-tumor effects of the constituent components of the antibody-drug conjugate and its formulation and to relate these effects to the anti-tumor effects of the intact immunoconjugate. Results: Mice were implanted by trocar with fragments of SK-ME-2 melanoma and, after the tumors became established, treatment with CROl l-vcMMAE and various components was tested to demonstrate the specificity of anti-tumor effects of this agent. Control groups, dosed with either the phosphate-buffered saline (vehicle) or the excipients of the immunoconjugate preparation (3% DMSO, sucrose, phosphate medium) steadily increased in tumor size to a maximum of 2,000 mg, at which time they were removed from the study. No apparent or statistically significant anti-tumor effects were observed. However, CROl l- vcMMAE treatment (at 5 mg/kg/treatment, q4d x4) produced measurable inhibition after the first 2 doses. Tumor growth inhibition continued until no discernible tumor was detected in all 6 of the test animals (Figure 4). In preliminary studies, tumor regression was complete and was not followed by regrowth of the tumor despite lengthy observation periods (up to 200 days). Conclusions:
The regressions produced by the immunoconjugate were not due to the individual components of the immunoconjugate nor to components of the formulation of that immunoconjugate. This is demonstrated by the lack of tumor growth inhibition after treatment with CROI l antibody alone (group 3) or free monomethylauristatin E (group 4), where the doses applied were identical to that contained in the intact immunoconjugate. Furthermore, the lack of anti-tumor effects noted with free MMAE suggests that anti-tumor effects from MMAE as a result of slow release from the antibody-drug conjugate may not explain the anti-tumor effects of the immunoconjugate. Release of MMAE from antibody- MMAE conjugates has been shown to be a very slow process in vivo (Ty,β = 6.0 days in the case of the anti-CD30 antibody- Auristatin E immunoconjugate (Sanderson et ai, Clin. Cancer Res. 11 : 843-852 (2005)) and would provide for plasma or serum concentrations that would be considerably lower than the "bolus" doses used in this study, which were ineffective at slowing the growth of the human melanoma xenografts.
Example 20: CROll-vcMMAE Inhibits the Growth of Human SK-MEL-5 Melanoma Xenografts Leading to Complete Regression of Established Melanoma Tumors in Athymic Mice (CROl l-ONC-3)
This study was performed to assess the potency and therapeutic efficacy of the antibody-drug conjugate, CROl l-vcMMAE, against a second model of established human melanoma, the SK-MEL-5 xenograft. Results: Though unrelated in origin, the SK-MEL-5 expresses GPNMB on the surface of the cell membrane and is killed by CROl l-vcMMAE in vitro. In this study, the anti-tumor effects of the CROI l immunoconjugate were examined, along with the vehicles PBS and saline, and the reference agents vinblastine and paclitaxel. In a manner similar to the SK- MEL-2 tumor, vinblastine produced a noticeable, but not significant tumor growth inhibition (P < 0.21) when compared to saline and PBS control groups (Figure 5). Soon after the commencement of treatment with paclitaxel, however, significant tumor growth inhibition was observed (P < 0.039) at day 3 after treatment began, and this anti-tumor effect continued, producing 100 % growth inhibition (stasis). The responses of SK-MEL-5- bearing test animals to vinblastine and paclitaxel were short-lived. After cessation of treatment at the maximally tolerated doses, tumors resumed rapid, progressive growth. One long-term, tumor-free survivor occurred in the paclitaxel group and one spontaneous regression occurred in the group treated with saline.
Substantial tumor growth inhibition, as well as tumor growth delay and complete regressions occurred in SK-MEL-5 tumor-bearing animals after treatment with CROI l- vcMMAE, and these effects were dose-related. At 10 mg/kg/treatment, significant antitumor effects were noted as early as 7 days (the equivalent of 2 treatments) after treatments began, when compared to saline (P < 0.0096), and as early as 10 days after treatment began when compared to PBS-treated controls (P = 0.039). In a dose-related manner, CROl l- vcMMAE produced tumor growth delay leading to complete regressions of established SK- MEL-5 melanoma xenografts (see tabular insert to Figure 5 for proportions of animals with complete regressions). Complete regressions occurred at CROl l-vcMMAE doses of 2.5 mg/kg/treatment, but not at 1.25 mg/kg/treatment.
As in previous studies, no indication of toxicity by the immunconjugate occurred in treated animals as evidenced by mortality of effects on body weight or weight gain. Conclusions:
CROl l-vcMMAE exerts substantial, dose-dependent anti-tumor effects against established xenografts of the SK-MEL-5 human melanoma. After just one or two treatments significant tumor growth inhibition is noted and which leads to long-term tumor-free survivors. Complete regressions occurred at doses of > 2.5 mg/kg i.v., q4d X4. Example 21 : Pharmacokinetics of CROll-vcMMAE (CROl 1-PK-lA)
The purpose of this study was to determine the stability of CRO 11 -vcMMAE in vivo after intravenous injection, the anticipated route of clinical administration. Materials & Methods. The CROl 1 antibody component of CRO 11 -vcMMAE was measured by a sandwich style enzyme-linked immunosorbent assay (ELISA) where serum was added to the wells of microtiter plates coated with the cognate antigen (GPNMB, CG56972-03) for the CROI l antibody, and the amount of human antibody were detected with an anti-globulin conjugated to the signal generator (horseradish peroxidase). Results:
Pharmacokinetics. The persistence of compound availability for antibody component of CROl 1 -vcMMAE was examined in a pharmacokinetic study in athymic mice (study CROl l-PK-1, Figure 6). The serum concentration-time profile for the antibody-drug conjugate was determined in athymic mice after intravenous administration of CROIl- vcMMAE and the results are presented in Figure 6. Athymic mice receiving 1 or 10 mg/kg intravenously showed dose-proportional serum concentrations over the entire span of sampling times (42 days). The concentration-time pattern was bi-phasic. The initial phase (α), however, was minor as it contributed <2% of the total AUC. Nevertheless, the compound disappeared very slowly from the peripheral blood (T>/2β = 10.3 days) with serum concentrations of 1 μg/mL and 10 μg/mL remaining in the blood for 6 weeks after dosing.
Estimates for the pharmacokinetic parameters for CROl 1 -vcMMAE are presented in Table 48. One parameter is noteworthy. The volume of distribution at steady state (Vss) is very low, approaching the theoretical minimum; this suggests that the corn-pound does not distribute outside the extravascular space. The distribution pattern, as well as the β- elimination phase for CRO 11 -vcMMAE are in good agreement with values obtained for antibodies in general (see Reviews by Mahmood and Green, Clin. Pharmacokinet 44: 331- 347 (2005); or Lobo et al J. Pharm. Sci. 93: 2645-2668 (2004)) and agree with values obtained for an antibody- Auristatin E immunoconjugate with comparable drug loading (Hamblett et al, Clin. Cancer Res. 10: 7063-7070 (2004)). Table 48. PK Parameters for CROl 1-vcMMAE after Intravenous Administration.
Figure imgf000099_0001
Abbreviations: A: Pre-exponential constant for alpha phase; Alpha: Exponential rate constant for alpha phase; AUC Total area under the curve from 0 to infinity; B: Pre- exponential constant for beta phase; Beta: Exponential rate constant for beta phase; Cl: Total or systemic clearance; CmaX: Maximum observed concentration; MRT: Mean residence time; Volume: Volume of central compartment; Vss: Steady-state volume of distribution. Estimates for pharmacokinetic parameters are presented in Table 48. One parameter is noteworthy. The volume of distribution steady state (Vss) is, approaching the theoretical minimum. These data suggest that the compound did not distribute outside the extravascular space. Taken together, these data are in good agreement with data on other immunoconjugates bearing the -vcMMAE cytotoxic moiety (see Hamblett et al, Clin. Cancer Res. 10: 7063-7070 (2004)). Conclusions: The CROl 1-vcMMAE antibody-drug conjugate has a serum-concentration profile which favors continuous exposure sufficient for disruption and eradication of melanoma xenografts. The immunoconjugate after i.v. administration has a sufficiently long half-life to ensure exposure of tumor cells for extended periods (Ty2β = 10.3 days), and may not require frequent dosing. The durability of CROl l-vcMMAE in vivo {e.g., athymic mice) is comparable to other Auristatin E immunoconjugates.
Example 22: The Schedule Dependency of the Anti-Tumor Effects of CROIl- vcMMAE (CROl 1-ONC-l)
The purpose of this study was to determine the extent to which the curative antitumor effects of the CROl 1 antibody-drug conjugate are dependent on the dosing regimen and, if possible, to determine the optimum dosing interval for this xenograft model. Materials and Methods:
The protocol for this study is presented in Table 49. To test the hypothesis that curative anti-tumor effects are influenced by the dosing schedule, the anti-tumor effects of CROl l-vcMMAE were measured at 5 different dosing intervals (i.e., 0, 1, 4, 8, and 16 days between treatments) and for each dosing interval 3 dosage levels were employed (i.e., cumulative doses of 2, 8, and 32 tng/kg); for each group, n= 6 athymic mice.
Nota bene: Please note that, although all 5 sets of groups in this experiment (e.g., groups 5, 6, and 7 represent one set and received 32, 8, and 2 mg/kg cumulative dose, respectively) received the same cumulative doses, the first set receiving the "bolus dose" is different from the other 4 sets. The Cmaχ for each group in the "'bolus" set was likely fourfold higher that the Cmax for the other 4 sets (see section on pharmacokinetics for dose- linearity after i.v. administration), since 4 sets of groups received 4 treatments, whereas the first set received only one "bolus" treatment (see column 7, Table 49 below).
TABLE 49. Protocol for the Dosing Interval Study (CROl l-PHM-2).
Group Treatment ROA Dose Regimen Dosing No. Cum.
Interval Treatments Dose
(mg/kg (days) (n) (mg/kg)
1 Phosphate Buffered Saline Bolus 0 1 N.A.
2 CR011-AE i.v. 32 Bolus 0 1 32
3 CR011-AE i.v. 8 Bolus 0 1 8
4 CR011-AE i.v. 2 Bolus 0 1 2
5 CR011-AE i.v. 8 qd x4 1 4 32
6 CR011-AE i.v. 2 qd x4 1 4 8
7 CR011 -AE i.v. 0.5 qd x4 1 4 2
8 CR011-AE I.V. 8 q4d x4 4 4 32
9 CR011-AE i.v. 2 q4d x4 4 4 8
10 CR011-AE i.v. 0.5 q4d x4 4 4 2
11 CR011-AE i.v. 8 q8d x4 8 4 32
12 CR011-AE i.v. 2 q8d x4 8 4 8
13 CR011-AE i.v. 0.5 q8d x4 8 4 2
14 CR011-AE i.v. 8 q16d x4 16 4 32
15 CR011-AE i.v. 2 q16d x4 16 4 8
16 CR011-AE i.v. 0.5 q16d x4 16 4 2
17 Excipients i.v. N.A. q16d x4 16 4 N.A.
Results:
For this study, the frequency of complete regressions with long-term tumor-free survivors was determined after 5 different dosing intervals were examined empirically (i.e., 0, 1, 4, 8, and 16 days between treatments). The aggregate responses for each set of groups, where a set is defined as 3 groups of graduated dosage levels but one dosage interval (groups 5, 6, and 7 represent 1 set, all of which were treated with a dosing interval of 1 day) are shown in Figure 7. The aggregate responses for test animals responding to CROI l- vcMMAE appear to suggest that bolus dosing and intervals of 1 day and 4 days provide a very slight advantage to the proportion of cures, compared to longer intervals, such as 8 days and 16 days between doses. However, this effect was not significant (P <0.2904). The data therefore suggest that the anti-tumor effects of CROll-vcMMAE in the SK-MEL-2 model are not schedule-dependent. This conclusion is strengthened by the fact that test animals in the bolus set (groups 2, 3, and 4), which were exposed to plasma concentrations approximately four-fold higher than any of the other groups, did not show any greater percentages of cured subjects.
The original design of this study was expanded to include an examination of the effects of various dosage levels. For each set, one group of animals received a cumulative dose of 8 mg/kg, which, from previous studies employing a dosing interval of 4 days, provided consistent therapeutic effects leading to long-term tumor-free animals. In addition, cumulative doses of 2 mg/kg and 32 mg/kg were employed.
The effects of dosage levels, in conjunction with various dosing intervals, are presented in Figure 8. Athymic mice receiving a cumulative dose of 32 mg/kg showed complete regressions in 100 % of each group, regardless of dosing interval; that is, a cumulative dose of 32 mg/kg is schedule-independent and represents a dose which is well above that sufficient for complete regressions in 100 % of the test animals (5 groups of 6 animals/group = 30 test animals). Animals receiving 8 mg/kg cumulative dose did not demonstrate schedule dependency and showed nearly the same proportions of complete regressions (i.e., 28/30 = 93%); Test animals receiving 2 mg/kg (cumulative dose), which was recognized in preliminary studies to be below the threshold for cures (using a standardized regimen of q4d X4) appeared to be schedule dependent, though this was not significant, and produced a much lower proportion of complete regressors (i.e., 13%). Conclusions:
The data from the dosing interval study suggests that the responses of SK-MEL-2 melanoma xenografts are not dependent on the schedule of administration of CROI l- vcMMAE. While no advantage could be shown for bolus dosing or regimens with low dosing intervals, there is the suggestion that, below a certain threshold cumulative dose, there may be some advantage to combining multiple treatments into a single bolus dose. Example 23: GPNMB Transcript Expression in Human Melanoma
GPNMB was recently shown to be expressed in glioblastoma and to mediate the in vitro and in vivo invasiveness of glioblastoma-derived tumor cells (see,e.g., Loging et al, Genome Res. 10:1393-1402 (2000); and Rich et al, J. Biol. Chem. 278:15951-15975 (2003)). To confirm and extend these findings to additional cancer types, we examined the expression of GPNMB transcripts in human cancer cell lines and tissues.
Material and Methods:
Total RNA was isolated using the RNeasy kit with a DNase digestion step (Qiagen Inc., Valencia CA). RT-PCR was performed using the One Step RT-PCR kit (Qiagen) as follows. RT : 50 0C for 45 min and 95 0C for 15 min for 1 cycle. PCR: 1 min at 95 0C, 1 min at 50 0C and 2 min at 72 0C for 30 cycles with final extension for 10 min at 72 0C. Products were separated on a 2% agarose/0.33% low melting point agarose gel and visualized by ethidium bromide staining. The integrity of each RNA sample was verified via RT-PCR with primers designed to amplify GAPDH. Specific primers (5'-3') used were: GPNMB: Forward-GAATTCAGAGTTAAACCTTGAG (SEQ ID NO: 327) Reverse-CAGGAATCTGATCTGTTACCAC (SEQ ID NO: 328) MART-I : Forward-CTGACCCTACAAGATGCCAAGAG (SEQ ID NO: 329)
Reverse- ATC ATGC ATTGC A AC ATTTATTGATGG AG (SEQ ID NO: 330) Tyrosinase: Forward-TTGGCAGATTGTCTGTAGCC (SEQ ID NO: 331) Reverse-AGGCATTGTGCATGCTGCTT (SEQ ID NO: 332) pMEL-17: Forward-TATTGAAAGTGCCGAGATCC (SEQ ID NO: 333) Reverse-TGCAAGGACCACAGCCATC (SEQ ID NO: 334)
RTQ-PCR analysis was performed with an ABI Prism 7700 Sequence Detection System using TaqMan reagents (PE Applied Biosystems, Foster City, CA). Equal quantities of normalized RNA's were used as a template in PCR reactions for 40 cycles with GPNMB- specific primers to obtain threshold cycle (CT) values. The following primers (5'-3') were used:
Forward-TCAATGGAACCTTCAGCCTTA (SEQ ID NO: 335) Reverse-GAAGGGGTGGGTTTTGAAG (SEQ ID NO: 336) Probe-TET-CTCACTGTGAAAGCTGCAGCACCAG -TAMRA (SEQ ID NO: 337) Result: Our transcript expression analysis indicated that GPNMB was strongly expressed in a high percentage of human metastatic melanoma samples. Using RTQ-PCR, GPNMB was found to be highly expressed (CT<27.0) in 5/7 melanoma cell lines and 5/5 melanoma clinical specimens examined (Table 50). In contrast, GPNMB was not expressed in a renal carcinoma cell line, TK-10, that was used as a negative control in our experiments. Table 50: GPNMB transcript expression in human melanoma cell lines and clinical specimens
Sample Details Expression*
Cell lines
UACC-62 Met. Melanoma 21.2
Ml 4 Met. Melanoma, amelanotic 22.2
SK-Mel-5 Met. Melanoma, axillary node 22.9
SlC-Mel-28 Met. Melanoma, skin 24.1
WM-266-4 Met. Melanoma, skin 24.5
A-375 Met. Melanoma, skin 29.0
LOXIMVl Met. Melanoma, amelanotic 30.9
TK-10 Renal cell carcinoma 40.0 Clinical specimens
#1 Met. Melanoma 26.6
#2 Melanoma 26.4
#3 Melanoma 26.9
#4 Met. Melanoma 24.1
#5 Met. Melanoma 25.3
* Threshold cycle (Cr) values from RTQ-PCR analysis. Met: Metastatic.
To extend these results, we investigated the expression of GPNMB in a panel of 17 melanoma cell lines via semi-quantitative RT-PCR (Table 51). The results show that GPNMB transcript is highly expressed in 15/17 melanoma cell lines, weakly expressed in 1/17 melanoma cell line (A-375), and not detectable in 1/17 melanoma cell line (LOXIMVI) nor in the control TK-IO. Table 51. RT-PCR analysis
Expression*
Cell line Annotation GPNMB MART-I Tyrosinase pMel-17
M 14 Met. Melanoma, amelanotic +++ +++ +++ +++
SK-Mel-5 Met. Melanoma, axillary node +++ +++ +++ +++
SK-Mel-28 Met. Melanoma, skin +++ +++ +++ +++
WM-266-4 Met. Melanoma, skin +++ +++ +++ +++
SK-Mel-2 Met. Melanoma, skin +++ +++ +++ +++
UACC-257 Met. Melanoma +++ +++ +++ +++
A2058 Met. Melanoma, lymph node +++ +++ +++ +++
G361 Met. Melanoma, skin +++ +++ +++ +++
HT- 144 Met. Melanoma, skin +++ +++ +++ +++
MEWO Met. Melanoma, lymph node +++ +++ +++ +++
SK-Mel-3 Met. Melanoma. Lymph node +++ +++ +++ +++
MALME-3M Met. Melanoma +++ +++ +++ +++
UACC-62 Met. Melanoma -H-+ +++ +++ -
SK-Mel-24 Met. Melanoma, lymph node +++ - +++ -
RPMI-7951 Met. Melanoma, lymph node +++ - + -
A-375 Met. Melanoma, skin + - - -
LOXIMVI Met. Melanoma, amelanotic _ _
TK-IO Renal cell carcinoma - - - -
*RT-PCR analysis: Strongly (+++), weakly (+) or not detectable (-). Met: Metastatic.
Furthermore, comparing the expression of GPNMB transcript to known melanoma/melanocyte-associated gene transcripts (MART-I, tyrosinase andpMEL-17) in the melanoma cell lines (Table 51) demonstrated strong expression of MART-I, tyrosinase and pMEL-17 in 13/17, 14/17 and 12/17 melanoma cell lines, respectively. Notably, 12/17 samples co-expressed high levels of GPNMB and all three melanoma/melanocyte- associated genes. Both LOXIMVI and TK-IO cell lines, which had undetectable GPNMB expression, also lacked expression of the three melanoma/melanocyte-associated genes examined.
Example 24: Growth-inhibitory Activity of CROll-vcMMAE is Dependent on GPNMB Expression Material and Methods:
Flow cytometry: Quantitative analysis of GPNMB expression on the cell surface of cell lines was determined by flow cytometry. Approximately 1 x 106 cells were harvested, washed and incubated with a saturating amount (10 μg/mL) of either CROl 1 or isotype- matched control antibody in staining buffer containing PBS (pH 7.4), 4% FBS and 0.1% NaN3 for 30 min on ice, followed by washing and staining with R-Phycoerythrin (PE)- conjugated goat-anti-human antibody (Jackson ImmunoResearch Laboratories, Inc, West Grove, PA) at 1 : 100 for 30 min on ice. Cells were fixed in 1% paraformaldehyde/PBS and examined on a Becton Dickinson FACSCalibur flow cytometer. Data analysis was performed with Becton Dickinson Cell Quest software version 3.3 and the geometric mean fluorescence intensity ratio (GMR) was determined for each cell type.
Internalization of cell surface bound antibodies was assessed by a modified flow cytometry procedure. In brief, cell suspensions were labeled with 10 μg/mL unconjugated or MMAE-conjugated CROl 1 for 30 min on ice. After washing cells, incubation was shifted to 37°C for 1 hr to allow internalization of bound antibodies. Cells that remained on ice (total surface bound) or that were incubated at 37°C (internalized) were stained with PE- conjugated goat-anti-human antibody at 1 : 100 for 30 min to detect CROl 1 retained on the cell surface. Labeled cells were analyzed by flow cytometry as described above. The percentage of antibody internalized was determined using the GMRs and the following formula: Percent internalized = Total surface bound (4°C) - Total surface bound (370C)/ Total surface bound (4°C) x 100
Immunoprecipitation and immnnoblot analysis: Cells were harvested and lysed on ice for 30 min in lysis buffer containing 1 % NP-40, 0.15 M NaCl, 0.02 M Tris-HCl, 10% glycerol, 0.01 M EDTA and complete protease inhibitor mixture (Roche Molecular Biochemicals, Indianapolis, IN). Supernatants were collected and the protein concentration was determined with the BCA Protein Assay Kit (Pierce, Rockford, IL). For immunoprecipitation, 2 μg of primary antibody was added into 0.5-1 mg of total cell lysates and incubated at 40C for 3 hrs, followed by incubation with protein-A-agarose (Amersham Biosciences, Upsala, Sweden) on ice for 2 hrs. The agarose beads were washed in ice-cold TBST (PBS with 0.1% Tween-20). Immunoprecipitates were recovered from supernatants after boiling in Laemmli sample buffer and centrifugation. For immunoblot analysis, total cell lysates (50 μg) or immunoprecipitates were resolved under reducing condition on 4-20% Tris-glycine gels (Invitrogen) and electrophoretically transferred to 0.45-μm PVDF membranes (Invitrogen). Membranes were blocked with 3% BSA (Sigma, St. Louis, MO) in TBST for 3 hrs and probed with rabbit anti-GPNMB polyclonal antibody (1 : 1000) for 3 hrs. Peroxidase-conjugated goat anti-rabbit IgG (H+L) secondary antibody (Jackson ImmunoResearch Labs) was added and incubated for 30 min. The membranes were washed in TBST and subjected to enhanced chemiluminescence (Amersham) following the manufacturer's protocol.
Clonogenic Assays: The growth-inhibitory activity of CROl 1-vcMMAE was determined by clonogenic assay. Cells were plated in 96-well plates and allowed to recover overnight. Unconj ugated CRO 11 , free MMAE, CRO 11 -vcMMAE or isotype-matched vcMMAE conjugated antibody at various concentrations was added to sub-confluent cell cultures and incubated for 4 days at 37°C. The cells were then transferred into 6-well plates and allowed to form colonies. Colonies were stained with Giemsa stain (Sigma) and counted. The surviving cell fractions were calculated based upon the ratio of the treated sample and the untreated control. The results were expressed as a percentage of control using GraphPad Prism Version 4 software. The IC50 was defined as the concentration resulting in a 50% reduction of colony formation compared to untreated control cultures.
Results:
To demonstrate that CROl 1 -vcMMAE growth-inhibitory activity is dependent on GPNMB expression, full-length GPNMB protein was ectopically expressed in HEK293 cells. Immunoblot (Figure 9A) and FACS (Figure 9B) analyses confirmed that GPNMB was expressed in GPNMB/plasmid transfected cells. Treatment of cells with CROl 1- vcMMAE, followed by clonogenic assay, demonstrated that GPNMB-expressing HEK293 cells were more sensitive to CROl l-vcMMAE-mediated growth-inhibition than were control cells devoid of GPNMB expression (Figure 9C).
To further verify our findings, GPNMB-expressing SK-Mel-2 cells were transfected with siRNA to specifically inhibit endogenous GPNMB expression. lmmunoblot and FACS analyses perfoπned 2 and 4 days after transfection demonstrated that total GPNMB (Figure 10A) and surface GPNMB (Figure 10B) protein levels were significantly reduced in SK-Mel-2 cells after the transfection when compared to the control transfectants. The amount of GPNMB expression was reduced for at least 7 days after transfection. Treatment of these cells with CROl 1-vcMMAE demonstrated that SK-Mel-2 cells were less sensitive to the growth-inhibitory activity of CROl 1-vcMMAE following siRNA-mediated GPNMB knockdown (Figure 10C). Taken together, these data indicate that the growth-inhibitory activity of CROl 1-vcMMAE required cell surface GPNMB expression. Example 25: Cell Cycle Arrest and Induction of Apoptosis by CROll-vcMMAE
To evaluate CROl l-vcMMAE's mechanism of growth inhibition, cell cycle analysis was performed.
Material and Methods:
The cell cycle effects of CRO 11 -vcMMAE were evaluated after treating cells in complete growth medium for 24 or 48 hr. Briefly, cells were pulsed at the indicated times with 30 μM of bromodeoxyuridine (BrdU, Sigma) for 30 min, harvested, fixed and permeabilized in methanol. Nascent DNA synthesis was detected by anti- bromodeoxyuridine-FITC (BD Biosciences, San Jose, CA) staining. Total DNA content was detected using propidium iodide (PI, Sigma). For apoptosis analysis, cells were treated as above and labeled with Annexin V-FITC followed by propidium iodide exclusion using the Annexin V-FITC Apoptosis Detection kit I (BD PharMingen, San Diego, CA) according to the manufacturer's protocols. Flow cytometry (as described in the previous Example) was used to assay both cell cycle and apoptosis studies.
Results: GPNMB-positive SK-Mel-2 cells or negative TK-10 control cells were treated with
CROl 1-vcMMAE for various lengths of time, followed by bromodeoxyuridine for 30 minutes to detect nascent DNA synthesis and finally, propidium iodide to detect total DNA content. DNA synthesis and cell cycle progression were determined by flow cytometry (Table 52). Table 52. Cell cycle analysis of CROl 1-vcMMAE treated cells
Treatment (ng/nιL) % G, % S-phase % G2M % Sub-G,
SK-Mel-2
24 hour
Untreated 55.2 30.0 9.9 0.5
CROI l (1000) 63.6 25.2 6.4 0.5
IgG2-VcMMAE (1000) 65.9 21.8 5.8 0.8
CROl 1-vcMMAE (100) 56.0 26.9 12.4 0.2
CROl 1-vcMMAE (1000) 43.7 20.0 28.5 1.1
TK-IO
24 hour
Untreated 39.7 43.7 7.0 0.5
CROI l (IOOO) 42.0 39.8 6.3 0.3
IgG2-VcMMAE (1000) 42.8 40.2 5.9 0.3
CROl l-vcMMAE (lOO) 51.1 35.1 4.5 0.7
CROl 1-vcMMAE (1000) 52.6 34.2 3.9 0.8
Cell cycle analysis was carried out by flow cytometry and the percentages of cells in each phase of cell cycle were determined by CellQuest Software (Becton Dickinson).
Exposure of GPNMB-positive cells to 1000 ng/mL CROl 1-vcMMAE, but not to isotype control IgG2-vcMMAE for 24 hrs, resulted in a decreased percentage (10 %) of cells in Gl and S-phase and an increased percentage (18.6 %) of cells in G2/M when compared to untreated cells. In contrast, CROl 1-vc MMAE did not affect the cycling of GPNMB-negative cells. At 48 hr after the treatment, CROl 1-vcMMAE further reduced the percentage (1 1 %) of cells in Gl and S-phase and increased the percentage (24 %) of cells in G2/M.
The increase in the sub-Gl population following CROl 1-vcMMAE treatment suggested the onset of apoptosis. To investigate this possibility, analysis of apoptosis using Annexin-V surface binding and loss of propidium iodide (PI) exclusion was performed. Our results demonstrated that 1000 ng/mL of CRO 11 -vcMMAE induced apoptosis specifically in GPNMB-expressing cells as indicated by an 11 % increase in mono-stained (Annexin-V+/PI-) cells following 48 hr of CROl l-vcMMAE treatment (Table 53). Table 53. Induction of apoptosis in human melanoma cells by CROl 1-vcMMAE
% AnnV7PI+ % AnnV+/PI+ % AnnV~/Pl" %
AnnV+/Pl' Treatment (ng/mL) UL UR LL
LR
SK-Mel-2
48 hour
Untreated 1.23 1.23 94.37
3.16
CROI l (1000) 0.36 0.45 94.45
4.74
IgG2-VcMMAE (1000) 0.17 0.51 95.93
3.39
CROl l-vcMMAE (lOO) 0.30 0.40 89.93
9.37
CROl 1-vcMMAE (1000) 2.08 2.02 82.08
13.83
TK-10
48 hour
Untreated 0.54 0.66 96.92
1.87
CROI l (IOOO) 0.83 0.34 98.27
0.55
IgG2-VcMMAE (1000) 0.62 0.95 97.09
1.33"
CROl 1-vcMMAE (100) 0.71 0.57 97.72
1.00
CROl 1-vcMMAE (1000) 0.86 0.83 97.75
0.56
Apoptosis analysis was carried out by flow cytometry and the percentages of cells in quadrants UL (upper left), UR (upper right), LL (lower left) and LR (lower right) were determined by CellQuest Software (Becton Dickinson). AnnV: Annexin V-FlTC and PI: Propidium iodide.
In addition, an increase in dual-stained (Annexin- V+/PI+) cells following CROI l- vcMMAE treatment indicated that the CROI l immunoconjugate enhanced cell death. Together, these results suggest that CROl l-vcMMAE selectively induced G2/M cell cycle arrest followed by apoptotic cell death.
Example 26: CROIl: A naked fully human IgGl for use in melanoma therapy exploiting the mechanism of antibody-dependent cellular cytotoxicity (ADCC)
Fully human monoclonal antibodies (mAb)-IgG2 to CG56972/GPNMB, an antigen predominantly found on the surface of melanoma and brain tumor cells, were generated. The naked CRO 11 IgG2 mAb (niAb 1.15) had no effect on CG56972 expressing cells either in vitro or in vivo. Thus we examined whether isotype switching from an IgG2 to an IgGl might enable the mAb to kill human melanoma cells through ADCC effector functions. Briefly, to switch CROl 1 from an IgG2 to IgGl antibody, double stranded DNA encoding constant region of IgGl (allotype Gm(f)) was synthesized, and IgG2 constant region was replaced with IgGl constant region using overlapped PCR approach. The sequences are described below: CRO 11 m Ab 1.15.1 mature heavy chain (IgG2) :
QVQLQESGPGLVKPSQTLSLTCTVSGGSISSFNYYWSWIRHHPGKGLEWIGYIYYSG STYSNPSLKSRVTISVDTSKNQFSLTLSSVTAADTAVYYCARGYNWNYFDYWGQG TLVTVSSASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNSGALTSGV HTFPAVLQSSGLYSLSSWTVPSSNFGTQTYTCNVDHKPSNTKVDKTVERKCCVEC PPCPAPPVAGPS VFLFPPKPKDTLMISRTPEVTCVWDVSHEDPEVQFNWYVDGVE VHNAKTKPREEQFNSTFRVVSVLTWHQDWLNGKEYKCKVSNKGLPAPIEKTISKT KGQPREPQVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTP PMLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID NO: 394) CRO 11 m Ab 1.15.1 mature heavy chain (IgG 1 ):
QVQLQESGPGLVKPSQTLSLTCTVSGGSISSFNYYWSWIRHHPGKGLEWIGYIYYSG STYSNPSLKSRVTISVDTSKNQFSLTLSSVTAADTAVYYCARGYNWNYFDYWGQG TLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGV HTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKRVEPKSCDKTH TCPPCP APELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDG VEVHNAKTKPREEQYNSTYRWSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTI SKAKGQPREPQVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYK TTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID NO: 395) We first analyzed the binding properties of the IgGl and IgG2 fully human monoclonal antibodies on SK-MEL-2 melanoma cells that have been shown to express CG56972 on the cell surface and bind CROl 1 IgG2. As shown in Figure 11, both the IgGl and IgG2 mAbs caused comparable FACS shifts on SK-MEL-2 cells compared to isotype control mAbs (Figure 11) indicating that both isotypes bind to CG56972/GPNMB with comparable saturation densities and affinities. We next examined whether the CROl 1 IgGl mAb could induce ADCC in SK-MEL-
2 cells in culture in the presence of human PBMC. Human PBMC were isolated from whole blood using a Ficoll-Plaque. Briefly, in a 50 mL tube, 15 mL of PBS was added to 20 mL of whole blood which was underlayed with 10 mL Ficoll-Plaque and the tube was centrifuged at 2000 RPM. Mononuclear cells were collected from the interface and washed
3 times with PBS. The ADCC assay was carried out in a 96 well plate using a fluorescence assay for cytolysis from Perkin-Elmer (DELFIA EuTDA Cytotoxic assay). The procedure is based on loading target cells with a fluorescence enhancing ligand (BATDA, bis (acetoxymethyl) terpyiϊdine - dicarboxylate). The hydrophic ligand penetrates the membrane quickly. Within the cell the esterbonds are hydrolyzed to form a hydrophilic ligand (TDA, terpyridine - dicarboxylic acid) which can no longer pass through the membrane. After cytolysis the ligand is released and introduced to the Europium solution. The europium and the ligand form a highly fluorescent and stable chelate (EuTDA). Fluorescence intensity are recorded using excitation and emission wavelengths as λex = 340 nm and λem = 613 nm, respectively.
Antibody-dependent cell-mediated cytotoxicity on SK-MEL-2 cells was assayed in the presence of PBMC and CROl 1 monoclonal antibody using effector: target ratios of 10, 30, 60 and 100 and various concentrations of IgGl or IgG2 mAb against CG56972/GPNMB (2, 5, 10 μg/200μl). Our data showed that between 30 to 100 fold PBMC, IgGl mAb caused cytolysis of SK-MEL-2 cells in a dose dependent manner (Figure 12A) whereas IgG2 mAb did not show any cytolysis (Figure 12B). Therefore, we conclude that CROl 1 IgGl mAb to CG56972/GPNMB can kill CG56972/GPNMB expressing melanoma cells in vitro and potentially human melanoma in vivo through ADCC effector functions. CROl 1 IgGl mAb can also be useful in combination with immune effector cytokines that could provide some clinical benefit in metastatic melanoma such as high dose IL-2, interferon- gamma or TNF-alpha. CROl 1 can also be used to treat melanoma in combination with vaccine immunotherapy, immunomodulators such as MDX-010, radiation therapy and/or chemotherapy.
Example 27: Treatment of Astrocytoma, Glioblastoma, Medulloblastoma and Other tumors of the CNS
Astrocytoma/glioblastoma is a highly drug-refractory neoplasm representing significant unmet medical needs. We identified CG56972 as a human gene (also known as GPNMB) that is highly expressed in these human cancer tissues and cancer cell lines. CG56972 is a type I transmembrane protein potentially involved in vesicular trafficking with a very restricted expression pattern in human brain. We generated fully human monoclonal antibodies against the CG56972 extracellular domain (amino acids 23-480). Our lead monoclonal antibody, designated CROl 1-vcMMAE was biochemically characterized and tested for therapeutic activity against cell lines derived from human brain tumors of astrocytoma, glioblastoma, medulloblastoma or neuroectodermal origin.
Transcript expression analysis demonstrated highly elevated CG56972 mRNA in brain tumors derived from astrocytoma, glioblastomas, medulloblastoma and tumors of neuroectodermal origin with restricted low expression in normal brain. CROl 1 bound by FACS analysis surface CG56972 on brain cancer cell lines. CROl 1 mAbs western blotted the predicted 100 and 120 kDa gene products. Clonogenic assays demonstrated that CROl 1-vcMMAE mAbs inhibited the growth of brain cancer cell lines.
Material and Methods:
Cell lines and culture conditions: All human cell lines, SK-MEL-2, XF-498, SNB- 78, U-118-MG, SF-539, H79MG, D392-MG, D534-MG, SK-N-SH, U-251 , SF-295, D450- MG, U87MG, SF-268, T98G, and SW-1783 were obtained from the American Type Culture Collection (Manassas, VA) or were purchased from the NCI (Bethesda, MD). Cells were maintained in DMEM or RPMI (Invitrogen, Carlsbad, CA) containing 10% FBS (Gemini Bio-Products, Woodland, CA) and penicillin-streptomycin. Real-Time Quantitative PCR (RTQ-PCR): Total RNA was isolated using the
RNeasy kit with a DNase digestion step (Qiagen Inc., Valencia). RNA samples were derived from normal human tissues obtained commercially (Clontech, Palo Alto, CA; Invitrogen, Carlsbad, CA) or cell lines grown according to specifications. RNAs were harvested and PCR was performed as previously described (Shimkets RA et. al. Nat Biotechnol., 1999. 17-8: 798-803) using TaqMan© reagents (PE Applied Biosystems, Foster City, CA). RNAs were normalized utilizing human β-actin and glyceraldehyde-3- phosphate dehydrogenase (GAPDH) TaqMan© probes according to the manufacturer's instructions. Equal quantities of normalized RNA were used as templates in PCR reactions with CG56972-specific reagents to obtain threshold cycle (CT) values. For graphic representation, CT numbers were converted to relative expression, relative to the sample exhibiting the highest level of expression. RTQ-PCR analysis was performed with an ABI Prism 7700 Sequence Detection System using TaqMan reagents (PE Applied Biosystems, Foster City, CA). The following primers (5 '-3') were used:
Forward-TCAATGGAACCTTCAGCCTTA (SEQ ID NO: 338) Reverse-GAAGGGGTGGGTTTTGAAG (SEQ ID NO: 339) Probe-TET-CTCACTGTGAAAGCTGCAGCACCAG -TAMRA (SEQ ID NO: 340)
CuraChip™: Tissues were lysed in Trizol. Biotin-labeled cDNA was made by using 15 mg of total RNA with poly(T) primers. Gene expression was evaluated by hybridization to the proprietary CuraChip microarray (CuraGen, New Haven, CT) of 11,000 oligonucleotide probes. Slides were hybridized for 15 h at 300C with constant rotation, washed for 30 min at room temperature (RT), incubated in streptavidin solution (4°C,
30min), washed three times for 15 min at RT, incubated in Cy3-conjugated detection buffer (4°C, 30 min), and washed three times for 15 min at RT. Slides were scanned (GMS 418 Scanner,Genetic Microsystems, Woburn, MA) and analyzed by using IMAGENE software (BioDiscovery, Marina Del Rey, CA). Data was subjected to 90th percentile normalization, and expression of the CG56972 gene was analyzed in comparison to that of the housekeeping gene GAPDH. The oligonucleotide sequence used to detect CG56972 is 5'- TGATCAGTAAGGATTTCACCTCTGTTTGTA (SEQ ID NO: 341). The oligonucleotide sequence used to detect GAPDH is 5'-ACCTTGTCATGTACCATCAATAAAGTACCC (SEQ ID NO: 342), corresponding to bp 1243-1272 of the GAPDH transcript (accession no. NM_002046).
Flow Cytometry: Quantitative analysis of CG56972 expression on the surface of cell lines was determined by flow cytometry (FACS). Approximately 1 x 106 cells were harvested, washed and incubated with a saturating amount (10 μg/mL). of either CROIl or isotype-matched control antibody in staining buffer containing PBS (pH 7.4), 4% FBS and 0.1% NaN3 for 30 min on ice, followed by washing and staining with R-Phycoerythrin (PE)-conjugated goat-anti-human antibody (Jackson ImmunoResearch Laboratories, Inc, West Grove, PA) at 1 :100 for 30 min on ice. Cells were fixed in 1% paraformaldehyde/PBS and examined on a Becton Dickinson FACSCalibur flow cytometer. Data analysis was performed with Becton Dickinson Cell Quest software version 3.3 and the geometric mean fluorescence intensity ratio (GMR) was determined for each cell type.
Immunoblot analysis: SK-MEL-2, XF-498, SNB-78, U-118-MG, SF-539, H79MG, D392-MG, D534-MG, SK-N-SH, U-251, SF-295, D450-MG, U87MG, SF-268, T98G, and
SW-1783 cells were harvested and lysed on ice for 30 min in lysis buffer containing 1 % NP-40, 0.15 M NaCl, 0.02 M Tris-HCl, 10% glycerol, 0.01 M EDTA and complete protease inhibitor mixture (Roche Molecular Biochemicals, Indianapolis, IN). Supernatants were collected and the protein concentration was determined with the BCA Protein Assay Kit (Pierce, Rockford, IL). For immunoblot analysis, 40 ul of total cell lysate from one well of confluent cells harvested from a 6 well Falcon tissue culture dish were boiled in Laemmli sample buffer, centrifuged and resolved under reducing condition on 4-20% Tris-glycine gels (Invitrogen). Gels were electrophoretically transferred to 0.45-μm PVDF membranes (Invitrogen). Membranes were blocked with 3% BSA (Sigma, St. Louis, MO) in TBST for 3 hrs and probed with goat anti-GPNMB polyclonal IgG (R & D Systems; 1 μg/mL, total 10 μg)) for 3 hrs. Peroxidase-conjugated anti-goat secondary antibody (Jackson
ImmunoResearch Labs) was added and incubated for 30 min. The membranes were washed in TBST and subjected to enhanced chemiluminescence (Amersham) following the manufacturer's protocol.
Clonogenic assays: The growth-inhibitory activity of CRO 11 -vcMMAE was determined by clonogenic assay. Cells were plated in 96-well plates and allowed to recover overnight. CROl 1-vcMMAE or isotype-matched monoclonal antibody at various concentrations was added to sub-confluent cell cultures and incubated for 4 days at 37°C. The cells were then transferred into 6-well plates and allowed to form colonies. Colonies were stained with Giemsa stain (Sigma) and counted. The surviving cell fractions were calculated based upon the ratio of the treated sample and the untreated control. The results were expressed as a percentage of control using GraphPad Prism Version 4 software. The IC50 was defined as the concentration resulting in a 50% reduction of colony formation compared to untreated control cultures.
Results: 1. CG56972 transcript expression in human astrocytoma, glioblastoma, medulloblastoma and tumors of neuroectodermal origin.
We examined the expression of CG56972 transcripts in human cancer cell lines and tissues (Figures 13 A & B). Our transcript expression analysis indicated that CG56972 was strongly expressed in all (15/15) human brain cancer cell lines tested (Figure 13A) Using RTQ-PCR, CG56972 was found to be expressed in cells of mixed glioblastoma/astrocytoma orgin, glioblastoma/gliomas, astrocytomas and metastatic neuroblastomas. The majority of brain or CNS tumor cell lines showed high level expression with CTs<27. Of note, CG56972 was found to be highly expressed (CT<27.0) in XF-498, U-118-MG, SNB-78, and SF-539 cells. As shown in Figure 13B, CG56972 was also expressed at high levels in 4/5 glioma human biopsies and 1/4 medulloblastoma human biopsies. Using microarray analysis from an in house chip containing a large panel of human genes (Figure 13C), CG56972 was found to be highly expressed in 5/9 brain cancers of astrocytoma or glioblastoma origin as well as 4/9 oligodendrogliomas. Our analysis of these tumor expression profiles showed that CG56972 message was detected to a much lesser degree in normal brain tissues. These data are also consistent with our immunohistochemical data that demonstrated the lack of CROl 1 staining in normal human brain including neurons and glial cells. Taken together, these data demonstrate that the CG56972 transcript is expressed at highly elevated quantities in brain cancer and oligodendroglioma cell lines and specimens isolated from human tumors.
2. Generation of fully human CROl 1 monoclonal antibodies to CG56972/GPNMB
The CG56972 protein is predicted to be a type I transmembrane glycoprotein. The highly elevated expression of CG56972 transcripts and the potential cell surface localization of this protein in human cancer samples encouraged us to generate monoclonal antibodies (mAbs) as a potential cancer therapeutic. Therefore, we cloned the human CG56972 extracellular domain (ECD; aa 23-480). Sequencing of the cloned cDNA revealed the presence of an in-frame 36-nt insertion, likely due to alternative splicing at the exon 6/7 boundary, which added an additional 12-aa (ATTLKS YDSNTP) (SEQ ID NO: 343) after residue 339 of the published GPNMB protein sequence. We verified the authenticity of 36-nt insertion via RT-PCR. The cDNA was next expressed in human HEK293 cells. The resultant protein was harvested, purified from the conditioned media and used as an immunogen to generate fully human mAbs against CG56972-ECD. Following immunization of XenoMouse©, mAbs that specifically recognized the CG56972- ECD protein via ELISA were generated. Our lead mAb, designated 1.15 or CRO 11 against purified CG56972-ECD, exhibiting a Kd of 52 nM against purified CG56972-ECD protein, was selected for in depth characterization and will be the focus of the remainder of this example. 3. CRO 11 mAb 1.15 detection of CG56972 protein expression in human brain cancers
We further used CROl 1 monoclonal antibodies to examine the surface expression of CG56972 protein on a variety of brain cancer cell lines by flow cytometry (Figure 14 & Table 54). FACS analysis demonstrated that the brain cancer cell lines XF- 498, U-1 18-MG, SNB-78 and SF-539; all positive for CG56972 transcript expression exhibited surface staining with CROl 1 monoclonal antibodies of at least 1.5-fold above isotype control mAb background (Figure 14). The cell line SF-268, that was the most weakly positive (CT>32) for the CG56972 transcript expression (Figure 13C & Table 54) showed minimal surface staining as expected of around 1.5-fold above control mAb background. Our SK-MEL-2 melanoma cell line that is our positive control for CG56972 expression showed strong cell surface staining.
To investigate CG56972 protein expression in our panel of brain cancer cell lines, total cell lysates were harvest, resolved by SDS-PAGE, transferred to membrane filters and subjected to immunoblot analysis with a CG56972 polyclonal antibody. As shown in Figure 15, the CG56972 polyclonal antibody detected two protein species that are differential glycosylation products of CG56972 of approximately 100 and 120 kDa from various brain cancer cell lines that have been shown to express CG56972 transcripts (Figure 13A). CG56972 protein was highly expressed in XP-498, SNB-78 and H79-MG and SF-539 cells. Both p 100 and p 120 species were detected to a lesser extent in U- 118-MG, U251 , D534-MG and D450-MG. Little or no CG56972 protein was detected in weakly expressing CG56972 transcript cell line, SF-268. An isotype-matched control IgG2 antibody did not immunoprecipitate CG56972 from any of the cell lines examined. All of these data are consistent with the cell surface expression of the CG56972 protein of the predicted molecule weights in brain cancer.
4. In vitro growth-inhibition of astrocytoma/glioblastoma cell lines with CRO 11 - vcMMAE.
CG56972 possesses a very restricted human tissue expression pattern. In preliminary studies, CROl 1 did not inhibit the growth of CG56972-expressing cancer cell lines when used directly (data not shown). Since CG56972 is a cell surface molecule on brain cancers and melanoma, and since CROl 1 was internalized following incubation with CG56972-expressing cells, we evaluated whether CROI l would inhibit the growth of cancer cells when combined with a protein synthesis inhibitor (saporin)-conjugated secondaiy antibody. Our results indicated that CROl 1 could specifically inhibit the growth of
CG56972-expressing cancer cells (data not shown). Thus, we conjugated CROl 1 directly to the cytotoxic drug monomethyl aurostatin E (MMAE) through a highly stable, but intracellular protease cleavable valine-citrulline (vc) linker. The resulting antibody-drug conjugate was named CROl 1-vc MMAE.
To investigate whether CROl 1-vcMMAE specifically inhibited the growth of brain cancer cells, clonogenic assays were performed to assess cell viability after CROl 1- vcMMAE treatment. As shown in Figure 16 and Table 54, our results indicated that CG56972-expressing cells were sensitive to growth-inhibition induced by CROl 1- vvMMAE, but not cells that poorly expressed this antigen (see SF-268 and LOXIMVI) at concentrations of vcMMAE less than 3 μg/mL. Strikingly, CROl 1-vcMMAE possessed IC50s of approximately 215, 450, 1250, and 1050 ng/mL on XF-498, SNB-78, U-118-MG and SF-539 cells (Figure 16 and Table 54). In these experiments, IC50s correlated with cell surface density as measured by FACS analysis. In contrast, conjugated control human IgG2 antibody- vcMMAE failed to inhibit the growth of any of the cell lines examined at concentrations up to 3 μg/mL (Table 54) with IC50s exceeding 1.5 or 4.5 μg/mL (Table 54).
Table 54. Summary of RTQ PCR, FACS and in vitro growth inhibition of human cancer cell lines with CRO 11 -mAbs
Figure imgf000117_0001
aCR011 vcMMAE ( mAb 1.15): CT values were determined by RTQ PCR as described in Materials and Methods. Geometric Mean ratios (GMR) were determined by flow cytometric analysis. Antibody-Drug
Cytotoxicity (ADC) or cell killing was determined by clonogenic assay as described. b IC50 value is the mean and SD of a representative clonogenic assay with each experiment performed in triplicate wells. ND: Not done. Conclusion:
These data indicate that CRO 1 1 -vcMMAE can be a highly potent and selective agent for the treatment of astrocytoma/glioblastoma and their metastasis as well as brain tumors of medulloblastoma and neuroectodermal origin. CROl 1-vcMMAE can also be useful for the treatment of melanoma metastasis to brain and other brain neoplasms such as neoplastic meningitis.
Example 28: Engineered Antibodies Derived from CROIl
The CROl 1 bi-scFv's (see Figure 17) of this work were designed to bind to a CD3 epitope of the T cell receptor on cytotoxic human T lymphocytes and, at the same time, to target diseased cells expressing GPNMB, with the net result of facilitating the lysis or destruction of the diseased cells.
The VL and VH domains of mAb CROIl, clone 1.15 were used in the construction of 3 CROl 1 based engineered antibodies:
(1) CROI l single chain antibody (CROI l scFv) (2) CROl 1 x anti-CD3 bispecific single chain antibody (bi-scFv), Linker set L4-L2-L4 (3) CROI l x anti-CD3 bispecific single chain antibody (bi-scFv), Linker set L4-L4-L4
The components of the CROl 1 scFv protein were: Signal Peptide-VL (CROl I)- Linker 4-VH (CROl 1)-Flag Tag. The components of the CROl 1 x anti-CD3 bi-scFv (Linker set L4-L2-L4) protein were: Signal Peptide- VL (CROl 1)-Linker 4-VH (CROl 1)-Linker 2-VH (anti-CD3)-Linker 4-VL (anti-CD3)-Flag Tag. The components of the CROl 1 x anti-CD3 bi-scFv (Linker set L4-L4-L4) protein were: Signal Peptide-VL (CROl 1)-Linker 4-VH (CROl 1)-Lmker 4-VH (anti-CD3)-Linker 4-VL (anti-CD3)-Flag Tag.
The various DNA components outlined above were used to generate the three CROl 1 engineered antibody products. The DNA components were synthesized by Blue Heron and cloned into commercially available plasmid vectors by methods familiar to those skilled in the art. These plasmids were then used in PCRs to combine the components, indicated in the 3 examples above, to generate engineered antibody inserts for expression vectors. In the host expression system examples practicing this invention described below, we have used CHOKl mammalian cells for the CROl 1 expression vectors, but expression is not limited to these cells; it will be recognized by those skilled in the art that the CROl 1 engineered antibodies of this invention can be expressed using other vectors, systems and cells, including but not limited to: pET vectors, inducible and constitutive promoters, and hosts may include E. coli, Bacillus species, yeast, including Pichia pastoήs or insect cells. Other expression hosts can also include various plant species and transgenic animals such as goats. SP (Signal Peptide): We incorporated a signal peptide in our constructs in order to express products that will be secreted. The signal peptide which was utilized for expression from CHO cells was derived from an immunoglobulin light chain leader peptide (Jirik et al. , 1986), or from the CR002 antibody (CuraGen).
Order of the bi-scFv components: The order of the antibody variable domains was fixed in both bi-scFv constructs as follows: VLI -L-VH I-L- VH2-L-VL2. Each of the 4 V domains was linked by a linker segment, L. VLI and VHI represent the immunoglobulin light and heavy chain variable domains respectively of CROl 1, and VH2 and VL2 represent the immunoglobulin heavy and light chain variable domains respectively of an anti-CD3 antibody that was used for both bi-scFv constructs. Other orders of the V domains can also be used for the 2 scFv components, as recognized by those skilled in the art, and the products evaluated for biological activity.
Tag: We used the 8 amino acid Flag tag at the C-terminus of the CROl 1 engineered antibodies to facilitate detection and purification of the products (Hickman et al, 2000).
Anti-CD3 scFv: The sequences of the VL and VH components of the anti-CD3 antibody used to generate the bi-scFv constructs may be found in me NCBI database under accession number CAE85148 (Lutterbuese et al.)
Linkers used in constructs: The sequence of L2, a short 5 amino acid linker that links the 2 monomer scFv components together in CROl 1 x anti-CD3 bi-scFv (L4-L2-L4 linker set) is G4S (Mack et al, 1995). L4 is a 25 amino acid linker based on the 205C linker (Denzin et al, 1991): LSADDAKKDAAKKDDAKKDDAKKDL (SEQ ID NO: 344) and is used in both of the CROl 1 bi-scFv species to link the CROl 1 VL and VH and the anti-CD 3 VH and VL. In the case of the CROl 1 x anti-CD3 bi-scFv (L4-L4-L4 linker set), L4 is also used to link the 2 monomer scFv components together. For the CROl 1 scFv, the L4 linker was used to link the two variable domains together. 1. DNA plasmid constructs for expression of CRO 11 scFv and CRO 11 X anti-CD3 bi- scFv species CROl 1 scFv Flag tag: The PCR amplification product for generating the expression construct for CROl 1 scFv was generated from a synthetic DNA template (Blue Heron) using the Fl/ Rl primers followed by nested PCR with the Fl nested/ Rl primer pair (Table 55) and Pfu Turbo DNA polymerase (Stratagene, cat# 600322), as per the manufacturer's directions. A Sal I/EcoR I PCR fragment coding for the CROl 1 scFv cassette was cloned into the corresponding restriction sites of the pCTN vector (CuraGen Corporation, mammalian expression vector) using the Fast-Link DNA Ligation kit (Epicentre, cat# LKl 1025).
Table 55
Figure imgf000120_0001
CROl 1 x anti-CD3 bi-scFv (L4-L2-L4) linker set Flag tag: The PCR amplification product for the CROl 1 x anti-CD3 bi-scFv having the (L4-L2-L4) linker set, was generated from a synthetic DNA template (Blue Heron) using the F2/ R2 primers followed by nested PCR with the F2 nested/ R2 primer pair (see Table 55 for sequences of oligonucleotides) and Pfu Turbo DNA polymerase (Stratagene, car# 600322), as per the manufacturer's directions. The Sal I/EcoR I PCR fragment having the coding sequence for the CRO 11 x anti-CD3 (L4- L2-L4) bi-scFv was cloned into the corresponding sites of the pCTN vector using Fast-Link DNA Ligation kit (Epicentre, cat# LKl 1025).
CROl 1 x anti-CD3 bi-scFv (L4-L4-L4) linker set Flag tag: The PCR amplification product for the CROl 1 x anti-CD3 bi-scFv having the (L4-L4-L4) linker set, was generated from a synthetic DNA template (Blue Heron) using the F3/ R3 primers followed by nested PCR with the F3 nested/ R3 primer pair (Table 55) and Pfu Turbo DNA polymerase (Stratagene, cat# 600322), as per the manufacturer's directions. The Nru 1/Xho I PCR fragment having the coding sequence for the CROl 1 x anti-CD3 (L4-L4-L4 linker set) bi-scFv was cloned into the corresponding sites of the pEE14.4FL2 expression vector (Lonza Biologies pic, 228 Bath Road, Slough, Berkshire SLl 4Dx, UK) using the Fast-Link DNA Ligation kit (Epicentre, cat# LKl 1025).
The DNA sequences of the above 3 expression construct inserts were verified by sequencing both strands of the relevant DNA products.
2. Protein Production of the CROl 1 Engineered Antibodies in CHOKl cells Adherent Chinese Hamster Ovary (CHOKl ) cells (ATCC catalog# CCL-61 ) were grown in DMEM media (Invitrogen, cat# 10564-011) supplemented with 10% fetal bovine serum (Gemini, cat#100106), GS supplement (JRH Biosciences, cat# 58672-100M) and 50 mg/L gentamicin (Invitrogen, cat# 15750078).
CHOKl cells were transfected with FuGENE 6 reagent (Roche, cat # 1815075) according to the manufacturer's directions. Expression and secretion was verified by Western blotting performed ca. 48 hours after the transfections. Selection of stable secreted CROl 1 scFv and CRO H x anti-CD3 bi-scFv (L4-L2-L4 linker set) lines were performed in selection media A (Table 56), while selection of a stable secreted CROI l x anti-CD3 bi-scFv (L4-L4- L4 linker set) line was performed in selection media B (Table 57). Table 56
Figure imgf000121_0001
Figure imgf000122_0001
In each case, 8 out of 96 CROl 1 scFv and CROl 1 x anti-CD3 (L4-L2-L4 linker set) bi- scFv CHOKl clones that were secreting products were expanded and archived. The best stable clones secreting products in each case were adapted to suspension culture in shake flasks with selection media C (Table 58) at 37°C and 5% CO2. Protein production for CROl lscFv and CROl lxCD3 (L4-L2-L4 linker set) bi-scFv was carried out in 4 L of selection media D (Table 59) at 300C and 5% CO2.
Table 58
Figure imgf000122_0002
Table 59
Figure imgf000123_0001
Only one out of two hundred CROl 1 x anti-CD3 (L4-L4-L4 linker set) bi-scFv CHOKl clones was found to produce a secreted product; this clone was expanded and archived. Protein production for the CROl 1 x anti-CD3 (L4-L4-L4 linker set) clone was carried out using a cell factory apparatus (Nunc, cat#l 64327), in selection media B (Table 57), 1 mM sodium butyrate (Sigma, cat# B5887) at 37°C and 10% CO2.
3. Protein Purification of the CRO 11 Engineered Antibodies
Protein purification for the CROl 1 scFv Flag and CROl 1 x anti-CD3 (L4-L2-L4 linker set) bi-scFv Flag was accomplished in three chromatography steps, including affinity, ion exchange and size exclusion chromatographies. For the purification of CRO H x anti-CD3 (L4-L4-L4 linker set) bi-scFv Flag protein, affinity and size exclusion chromatographies were used.
Affinity chromatography was performed using anti-FLAG M2 affinity gel (Sigma, cat# A2220-25 mL) as per the manufacturer's instructions on a BioCAD 700E instrument (Applied Biosystems). Ion exchange chromatography was performed on a MonoQ 5/50 GL column (Amersham, cat# 17-5166-01) using 20 mM Tris-HCl pH7.5 as equilibration buffer and a gradient elution with 0 - 1 M NaCl. Size exclusion chromatography was performed using a Superdex 75/10/300 GL column (Amersham, cat#l 7-5174-01) following the manufacturer's protocols on BioCAD 700E (Applied Biosystems) liquid chromatography instrument.
The approximate yields from 1 L conditioned CHOKl media were: (1) CROI l scFv: 1.0 mg
(2) CROl 1 x anti-CD3 (L4-L2-L4 linker set) bi-scFv: 0.5 mg
(3) CROI l x anti-CD3 (L4-L4-L4 linker set) bi-scFv: 1.5 mg
The N-terminal amino acid sequence of the purified proteins was determined by Edman degradation, using methods known to those skilled in the art. The sequence of the first five amino acids was: E I V M T in each case (the mature N-terminus of the CROl 1 VL protein), indicating that accurate processing by signal peptidase had occurred to give a soluble, secreted product of the predicted sequence and size.
The DNA and amino acid sequences of the 3 CROl 1 engineered products are given below.
SEQ ID for CROl 1 scFv - (VL-L4-VH) Flag. The Signal peptide of Human kappa light chain was used as described in Kabat et al 45 CLL-CL). There was a FLAG tag included at the C-terminus. The Kozak sequence CCACC was included in the 5' PCR primer.
ATGGAAGCCCCAGCGCAGCTTCTCTTCCTCCTGCTACTCTGGCTCCCAGATACCACCGGTGAAAT AGTGATGACGCAGTCTCCAGCCACCCTGTCTGTGTCTCCAGGGGAAAGAGCCACCCTCTCCTGCA GGGCCAGTCAGAGTGTTGACAACAACTTAGTCTGGTACCAGCAGAAACCTGGCCAGGCTCCCAG GCTCCTCATCTATGGTGCATCCACCAGGGCCACTGGTATCCCAGCCAGGTTCAGTGGCAGTGGGT CTGGGACAGAGTTCACTCTCACCATCAGTAGTCTGCAGTCTGAAGATTTTGCAGTTTATTACTGTC AGCAGTATAATAACTGGCCTCCGTGGACGTTCGGCCAAGGGACCAAGGTGGAAATCAAACTTTC CGCGGACGATGCGAAAAAGGATGCTGCGAAGAAAGATGACGCTAAGAAAGACGATGCTAAAAA GGACCTGCAGGTGCAGCTGCAGGAGTCGGGCCCAGGACTGGTGAAGCCTTCACAGACCCTGTCC CTCACCTGCACTGTCTCTGGTGGCTCCATCAGCAGTTTTAATTACTACTGGAGCTGGATCCGCCAC CACCCAGGGAAGGGCCTGGAGTGGATTGGGTACATCTATTACAGTGGGAGCACCTACTCCAACC CGTCCCTCAAGAGTCGAGTTACCATATCAGTAGACACGTCTAAGAACCAGTTCTCCCTGACGCTG AGCTCTGTGACTGCCGCGGACACGGCCGTGTATTACTGTGCGAGAGGGTATAACTGGAACTACTT TGACTACTGGGGCCAGGGAACCCTGGTCACCGTCTCCTCAGCTAGCGATTATAAGGACGATGAT GACAAATAGTAA (SEQ ID NO: 354)
MEAPAQLLFLLLLWLPDTTGEIVMTQSP ATLSVSPGERATLSCRASQSVDNNLVWYQQKPGQAPRLL IYGASTRATGIPARFSGSGSGTEFTLT[SSLQSEDFAVYYCQQYNNWPPWTFGQGTKVEIKLSADDAK KDAAKICDDAKKDDAKKDLQVQLQESGPGLVKPSQTLSLTCTVSGGSISSFNYYWSWIRHHPGKGLE WIGYIYYSGSTYSNPSLKSRVTISVDTSKNQFSLTLSSVTAADTA VYYCARGYN WNYFDYWGQGTLV TVSSASDYKDDDDK (SEQ ID NO: 355)
SEQ ID for CROl 1 x anti-CD3 (L4-L2-L4 linker set) bi-scFv - The Signal peptide of Human kappa light chain was used as described in Kabat et al. 45 CLL-CL). There was a FLAG tag included at the C-terminus. ATGGAAGCCCCAGCGCAGCTTCTCTTCCTCCTGCTACTCTGGCTCCCAGATACCACCGGTGAAAT AGTGATGACGCAGTCTCCAGCCACCCTGTCTGTGTCTCCAGGGGAAAGAGCCACCCTCTCCTGCA GGGCCAGTCAGAGTGTTGACAACAACTTAGTCTGGTACCAGCAGAAACCTGGCCAGGCTCCCAG GCTCCTCATCTATGGTGCATCCACCAGGGCCACTGGTATCCCAGCCAGGTTCAGTGGCAGTGGGT CTGGGACAGAGTTCACTCTCACCATCAGTAGTCTGCAGTCTGAAGATTTTGCAGTTTATTACTGTC AGCAGTATAATAACTGGCCTCCGTGGACGTTCGGCCAAGGGACCAAGGTGGAAATCAAACTTTC CGCGGACGATGCGAAAAAGGATGCTGCGAAGAAAGATGACGCTAAGAAAGACGATGCTAAAAA GGACCTGCAGGTGCAGCTGCAGGAGTCGGGCCCAGGACTGGTGAAGCCTTCACAGACCCTGTCC CTCACCTGCACTGTCTCTGGTGGCTCCATCAGCAGTTTTAATTACTACTGGAGCTGGATCCGCCAC CACCCAGGGAAGGGCCTGGAGTGGATTGGGTACATCTATTACAGTGGGAGCACCTACTCCAACC CGTCCCTCAAGAGTCGAGTTACCATATCAGTAGACACGTCTAAGAACCAGTTCTCCCTGACGCTG AGCTCTGTGACTGCCGCGGACACGGCCGTGTATTACTGTGCGAGAGGGTATAACTGGAACTACTT TGACTACTGGGGCCAGGGAACCCTGGTCACCGTCTCCTCAGGAGGTGGTGGATCCGATATCAAA CTGCAGCAGTCAGGGGCTGAACTGGCAAGACCTGGGGCCTCAGTGAAGATGTCCTGCAAGACTT CTGGCTACACCTTTACTAGGTACACGATGCACTGGGTAAAACAGAGGCCTGGACAGGGTCTGGA ATGGATTGGATACATTAATCCTAGCCGTGGTTATACTAATTACAATCAGAAGTTCAAGGACAAGG CCACATTGACTACAGACAAATCCTCCAGCACAGCCTACATGCAACTGAGCAGCCTGACATCTGA GGACTCTGCAGTCTATTACTGTGCAAGATATTATGATGATCATTACTGCCTTGACTACTGGGGCC AAGGCACCACTCTCACAGTCTCCTCACTTTCCGCGGACGATGCGAAAAAGGATGCTGCGAAGAA AGATGACGCTAAGAAAGACGATGCTAAAAAGGACCTGGACATTCAGCTGACCCAGTCTCCAGCA ATCATGTCTGCATCTCCAGGGGAGAAGGTCACCATGACCTGCAGAGCCAGTTCAAGTGTAAGTT ACATGAACTGGTACCAGCAGAAGTCAGGCACCTCCCCCAAAAGATGGATTTATGACACATCCAA AGTGGCTTCTGGAGTCCCTTATCGCTTCAGTGGCAGTGGGTCTGGGACCTCATACTCTCTCACAA TCAGCAGCATGGAGGCTGAAGATGCTGCCACTTATTACTGCCAACAGTGGAGTAGTAACCCGCT CACGTTCGGTGCTGGGACCAAGCTGGAGCTGAAAGCTAGCGATTATAAGGACGATGATGACAAA TAGTAA (SEQ ID NO: 356)
MEAPAQLLFLLLLWLPDTTGEIVMTQSP ATLSVSPGERATLSCRASQSVDNNLVWYQQKPGQAPRLL IYGASTRATGIP ARFSGSGSGTEFTLTISSLQSEDFA VYYCQQYNNWPPWTFGQGTKVEIKLSADDAK KDAAKKDDAKKDDAKKDLQVQLQESGPGLVKPSQTLSLTCTVSGGSISSFNYYWSWIRHHPGKGLE wiGYiYYSGSTYSNPSLKSRVTisvDTSKNQFSLTLSsvTAADTA VΎYCARGYNWNYFDYWGQGTLV
TVSSGGGGSDIKLQQSGAELARPGASVKMSCKTSGYTFTRYTMHWVKQRPGQGLEWIGYINPSRGY TN YNQICFKDKATLTTDKSSSTA YMQLSSLTSEDSA VYYCARYYDDHYCLDYWGQGTTLTVSSLSAD DAKKDAAKKDDAKKDDAKKDLDIQLTQSPAIMSASPGEKVTMTCRASSSVSYMNWYQQKSGTSPK RWIYDTSKVASGVPYRFSGSGSGTSYSLTISSMEAEDAATYYCQQWSSNPLTFGAGTKLELKASDYK DDDDK (SEQ ID NO: 357)
SEQ ID for CROl 1 x anti-CD3 (L4-L4-L4 linker set) bi-scFv - The Signal peptide of CR002 was used. There was a FLAG tag included at the C-terminus. ATGGAAACCCCAGCGCAGCTTCTCTTCCTCCTGCTACTCTGGCTCCCAGATACCACCGGAGAAAT AGTGATGACGCAGTCTCCAGCCACCCTGTCTGTGTCTCCAGGGGAAAGAGCCACCCTCTCCTGCA GGGCCAGTCAGAGTGTTGACAACAACTTAGTCTGGTACCAGCAGAAACCTGGCCAGGCTCCCAG GCTCCTCATCTATGGTGCATCCACCAGGGCCACTGGTATCCCAGCCAGGTTCAGTGGCAGTGGGT CTGGGACAGAGTTCACTCTCACCATCAGTAGTCTGCAGTCTGAAGATTTTGCAGTTTATTACTGTC AGCAGTATAATAACTGGCCTCCGTGGACGTTCGGCCAAGGGACCAAGGTGGAAATCAAACTTTC CGCGGACGATGCGAAAAAGGATGCTGCGAAGAAAGATGACGCTAAGAAAGACGATGCTAAAAA GGACCTGCAGGTGCAGCTGCAGGAGTCGGGCCCAGGACTGGTGAAGCCTTCACAGACCCTGTCC CTCACCTGCACTGTCTCTGGTGGCTCCATCAGCAGTTTTAATTACTACTGGAGCTGGATCCGCCAC CACCCAGGGAAGGGCCTGGAGTGGATTGGGTACATCTATTACAGTGGGAGCACCTACTCCAACC CGTCCCTCAAGAGTCGAGTTACCATATCAGTAGACACGTCTAAGAACCAGTTCTCCCTGACGCTG AGCTCTGTGACTGCCGCGGACACGGCCGTGTATTACTGTGCGAGAGGGTATAACTGGAACTACTT TGACTACTGGGGCCAGGGAACCCTGGTCACCGTCTCCTCATTATCAGCGGATGACGCCAAGAAA GACGCAGCCAAAAAGGACGATGCAAAGAAGGATGACGCAAAGAAAGATTTAGATATCAAACTG CAGCAGTCAGGGGCTGAACTGGCAAGACCTGGGGCCTCAGTGAAGATGTCCTGCAAGACTTCTG GCTACACCTTTACTAGGTACACGATGCACTGGGTAAAACAGAGGCCTGGACAGGGTCTGGAATG GATTGGATACATTAATCCTAGCCGTGGTTATACTAATTACAATCAGAAGTTCAAGGACAAGGCCA CATTGACTACAGACAAATCCTCCAGCACAGCCTACATGCAACTGAGCAGCCTGACATCTGAGGA CTCTGCAGTCTATTACTGTGCAAGATATTATGATGATCATTACTGCCTTGACTACTGGGGCCAAG GCACCACTCTCACAGTCTCCTCACTTTCCGCGGACGATGCGAAAAAGGATGCTGCGAAGAAAGA TGACGCTAAGAAAGACGATGCTAAAAAGGACCTGGACATTCAGCTGACCCAGTCTCCAGCAATC ATGTCTGCATCTCCAGGGGAGAAGGTCACCATGACCTGCAGAGCCAGTTCAAGTGTAAGTTACA TGAACTGGTACCAGCAGAAGTCAGGCACCTCCCCCAAAAGATGGATTTATGACACATCCAAAGT GGCTTCTGGAGTCCCTTATCGCTTCAGTGGCAGTGGGTCTGGGACCTCATACTCTCTCACAATCA GCAGCATGGAGGCTGAAGATGCTGCCACTTATTACTGCCAACAGTGGAGTAGTAACCCGCTCAC GTTCGGTGCTGGGACCAAGCTGGAGCTGAAAGATTATAAGGACGATGATGACAAATAGCTCGAG CGG (SEQ ID NO: 358)
METPAQLLFLLLLWLPDTTGEIVMTQSPATLSVSPGERATLSCRASQSVDNNLVWYQQKPGQAPRLLIY GASTRATGIPARFSGSGSGTEFTLTISSLQSEDFA VYYCQQ YNNWPPWTFGQGTKVEIKLSADDAKKDA AKKDDAKKDDAKKDLQVQLQESGPGLVKPSQTLSLTCTVSGGSISSFNYYWSWIRHHPGKGLEWIGYI YYSGSTYSNPSLKSRVTISVDTSKNQFSLTLSSVTAADTAVYYCARGYN WNYFDYWGQGTLVTVSSLS ADDAKKD AAKKDDAKKDDAKKDLDIKLQQSGAELARPGASVKMSCKTSGYTFTRYTMHWVKQRPG QGLEW[GYINPSRGYTNYNQKFKDKATLTTDKSSSTAYMQLSSLTSEDSAVYYCARYYDDHYCLDYW GQGTTLTVSSLSADDAICKDAAKKDDAKKDDAKKDLDIQLTQSPA[MSASPGEKVTMTCRASSSVSYM NWYQQKSGTSPKRWIYDTSKVASGVPYRFSGSGSGTSYSLTISSMEAEDAATYYCQQWSSNPLTFGAG TKLELKDYKDDDDK (SEQ ID NO: 359)
4. Testing of the 3 CROl 1 engineered antibodies by ELISA, flow cytometry and determination of cytotoxicity: ELISA : The binding of the CROl 1 engineered antibodies to purified recombinant GPNMB (2 μg/mL) was measured using plates coated overnight at 40C. Plates were then blocked and washed. Various dilutions of the CROl 1 engineered antibodies were added into the wells. Plates were incubated for 1 h and washed. HRP-conjugated anti-FLAG M2 mAb (Sigma, St. Louis, MO) was added into the wells for 1 h, washed and the reaction developed with the TMB substrate reagent as described by the manufacturer (Pharmingen, San Jose, CA).
Binding of the CROl 1 scFv and CROl 1 x anti-CD3 (L4-L2-L4 linker set) bi-scFv product to GPNMB was first confirmed using ELISA, as shown in Figure 18. Plates were coated with human GPNMB protein tagged with His and V5. Coated plates were incubated with either supernatants containing CROl 1 X anti-CD3 bi-scFv or purified CROl 1 scFv monomer. Binding of the recombinant mAbs (both monomer and dimer) was detected using anti-FLAG-HRP conjugated mAb M2 (Sigma). As can be seen in Figure 18, both anti-GPNMB antibody species described bind to the recombinant GPNMB protein, indicating that the specificity and binding activity of the engineered anti-GPNMB antibody, using the methods described in this example, was preserved.
Flow cytometry: The binding of the CROl 1 engineered antibodies to native proteins was analyzed by FACS. Briefly, human T cells and SK-Mel-5 cells were incubated with either the CROl 1 scFv or CROl 1 x anti-CD3 (L4-L2-L4 linker set) bi-scFv (5 μg/sample/100 μl) with subsequent staining with mouse anti-FLAG mAb (Sigma) and PE- conjugated goat anti-mouse Ig F(ab')2. (Jackson ImmunoResearch, West Grove, PA) Ten thousand events were collected and analyzed on a FACSCalibur instrument (Becton Dickinson, Mountain View, CA).
To confirm binding of the CROl 1 scFv and CROl 1 x anti-CD3 (L4-L2-L4 linker set) bi-scFv products to native GPNMB protein expressed on the cell surface, we used SK-MeI- 5 cells which naturally express GPNMB. To verify binding of the bi-scFv to human CD3 molecules, we used purified human T cells. As a positive control we used native PE conjugated anti-CD3-PE and CROl 1 mAb. Binding of the CROl 1 scFv was detected using anti-FLAG mAb M2 with subsequent staining with PE conjugated anti mouse IgG, while for detection of CROl 1 mAb binding we used anti-human IgG-PE. Control anti-CD3 mAb bound to T cells, and control anti-GPNMB mAb bound to SK-Mel-5 tumor cells. We found that only the CROl 1 x anti-CD3 (L4-L2-L4 linker set) bi-scFv stained T cells; the CROl 1 scFv monomer did not bind CD3 positive T cells, as expected (see Figure 19). Binding to SK-Mel-5 cells by either the CROl 1 scFv monomer or CROl 1 X anti-CD3 (L4-L2-L4 linker set) bi-scFv was present at a low level (Figure 19).
Cytotoxicity: The ability of CROl 1 x anti-CD3 (L4-L2-L4 linker set) bi-scFv to redirect human T lymphocytes to kill relevant human tumor cells was measured by flow cytometry. Tumor cells were labeled with PKH2 green fluorescent linker kit (Sigma) and washed. Purified T cells were cultured O/N with PKH2 -labeled tumor cells in the presence or absence of purified bi-scFv. Death of GPNMB positive tumor cells was measured by propidium iodine (PI) incorporation. To evaluate the ability of the CROl 1 x anti-CD3 (L4-L2-L4 linker set) bi-scFv product to increase T cell mediated killing of GPNMB positive cells, we performed a cytotoxicity test. Purified T cells were cultured O/N with PKH2-labeled SK-Mel-5 (GPNMB positive) tumor cells in the presence of various doses of purified CROl 1 scFv and CROl 1 x anti-CD3 (L4-L2-L4 linker set) bi-scFv products. Conclusion:
The CROl 1 x anti-CD3 (L4-L2-L4 linker set) bi-scFv significantly increased killing of SK-Mel-5 tumor cells by T lymphocytes (Figure 20). In contrast, the addition of monospecific anti-GPNMB scFv did not increase killing of SK-Mel-5 tumors. In addition, no cytotoxicity was observed when the tumor cells were cultured with the CROl 1 x anti-CD3 (L4-L2-L4 linker set) bi-scFv without T lymphocytes (Figure 20). These data indicate that the CROl 1 x anti-CD3 (L4-L2-L4 linker set) bi-scFv provided sufficient bridging between T cells and SK-Mel-5 cells to induce cell death and that both components of this engineered CROl 1 bi-specific antibody were biologically active. Therefore the CROl 1 x anti-CD3 (L4- L2-L4 linker set) bi-scFv engineered antibody of the present invention may be used as a therapeutic to treat diseases, such as melanoma and other cancers where there are upregulated levels of GPNMB and T cells present.
Other methods of cytotoxicity analysis, including fluorescence and chromium release assays can be used to demonstrate the usefulness of the CROl 1 x anti-CD3 (L4-L2- L4 linker set) bi-scFv in treating tumors. Other linkers may also be used to link the two scFv monomer components together, as in the CROl lxanti-CD3 (linker set L4-L4-L4) molecule described supra. Example 29: Optimized Production Process of CROll-vcMMAE
CROl IAE is an antibody-drug conjugate composed of the anti-GPNMB (CG56972) fully human antibody CROl 1 conjugated with the toxin Auristatin E through a protease- cleavable linker. The toxin-to-antibody ratio is approx. 4.0 but may vary between 3.5 and 4.2. While the CROl 1 antibody is IgG2, it is therefore possible to append up to 12 toxin molecules per antibody molecule using the free thiols as a reactive site.
The structure of Maleimidocoaproyl-Valine-Citrullin-Monomethyl-Auristatin E (vcMMAE) is shown in Figure 21.
Conjugation: A process of generating the drug-substance consisting of CROl 1 mAb with VCMMAE attached. An overview of the conjugation process is summarized in Figure
22.
Briefly, the conjugation process for CROl 1 fully human antibody consists of the following 4 steps. 1) Buffer exchange and sucrose removal by diafiltration, 2) Disulfides reduction, 3) Conjugation to vcMMAE and finally, 4) Purification of conjugated CROl 1- vc MMAE by diafiltration. There are several assays throughout the process, i.e. in-process assays, which include Ellman's assay and determination of protein concentration. At the end of the process, the drug substance, i.e. the conjugate, is analyzed for drug-to-antibody ratio, free drug content and protein concentration.
Diafiltration of the bulk antibody: The bulk antibody originally formulated in phosphate pH 7 -10% sucrose was buffer exchanged into the conjugation buffer (borate pH 9.0 - NaCl) by diafiltration over 10 diavolumes. At the end of diafiltration, CROl 1 was diluted to ~5.5 mg/niL and filtered through a set of two filters consisting of 1.2 and 0.22 μm. The buffer exchange is required because sucrose interferes with reduction. In addition, high pH improves CROl 1 solubility. CROIl reduction - General considerations: CROl 1 is produced as an IgG2 isotype product and contains 6 disulfide bridges in the hinge region. These disulfides can be reduced under mild conditions to give rise to 12 cysteine residues. Therefore, it is possible to maximally attach 12 vcMMAE drug molecules per antibody. For the process, however, the bulk antibody is only partially reduced because the aim is to generate conjugates with an average of 4 vcMMAE molecules. The reason for this is two-fold. First, it broadens the therapeutic window by decreasing potential systemic toxicity associated with MMAE. Second, it is difficult and sometimes impossible to produce fully-loaded conjugates with low aggregation because of greatly reduced solubility imparted by the hydrophobic drug.
Process: Tris-(carboxyethyl)-phosphine or TCEP was added at the 4:1 molar ratio (TCEP:mAb) to CROl 1 at a concentration of ~5.5 mg/mL in the jacketed reactor equipped with an agitator set to 90 RPM. The reaction was allowed to proceed for 3 hours at 37°C in the presence of 1 mM EDTA. At the end, Ellman's assay was used to determine the amount of free thiols. Typically, it was 4.2 thiols per antibody. The reactor was then chilled to 4°C.
CROl 1 conjugation - General considerations: TCEP was not fully consumed during the reduction. The left over TCEP was capable of reacting with vcMMAE. However, this spurious side reaction was slower compared to the conjugation reaction and can be mitigated by adding an excess of vcMMAE. The advantage of TCEP compared to DTT is that it does not require removal of the left-over reducing agent.
Process: vcMMAE was dissolved in DMSO and added at 20% molar excess to the reduced CROl 1 mAb. The reaction was allowed to proceed for 1 hour. The final concentration of DMSO is 4% (v/v). DMSO played a dual purpose in the process. It is required for solubilizing the drug and also it helps to solubilize the conjugate. At the end of conjugation, N-acetylcysteine was added to quench any unreacted drug.
CROl 1 -vcMMAE purification: The temperature in the reactor was brought to room temperature. A 40% sucrose stock solution was used to adjust the final sucrose concentration to 10% (w/v) followed by a pH adjustment using 300 mM histidine HCl pH 5.0 buffer to a final pH of 6.0. The conjugate was then purified by diafϊltration into 20 mM histidine pH 6.0-10% sucrose (w/v) buffer and using 10 diavolumes. At the end of diafiltration, the conjugate was concentrated to ~7 mg/mL and filtered through a set of three filters consisting of 1.2, 0.45 and finally, 0.22 μm. CROl 1 -vcMMAE formulation: The conjugate was formulated by adding Tween-20 to a final concentration of 0.02% and by diluting to 6 mg/mL (±10%) using formulation buffer (20 mM histidine pH 6.0, 10% sucrose, 0.02% Tween-20). The conjugate was then stored at 4°C until pooling if more than one lot is being manufactured (a.k.a. staging time). After pooling, the final concentration was adjusted to 5.0 mg/mL (±5%) and the drug substance was stored frozen. 1. Pre-conjugation UF/DF: Removal of Sucrose
Experiments were performed to monitor the rate of removal of sucrose during UF/DF by Ellman's assay; and estimate the diavolumes needed to achieve the highest SH- per-Ab ratio. It was found that it is desirable to conduct at least 6 diavolumes in order to remove sucrose to a level that does not impede CROl 1 reduction. To ensure robustness, at least 10 diavolumes should be utilized during the process.
2. The Effect of Percent DMSO on Aggregation in the Conjugation Reaction
Experiments were performed to determine the effect of DMSO in the conjugation reaction on: (1) aggregation; and (2) drug:Ab molar ratio {i.e. completeness of conjugation).
It was found that the percent aggregate in reaction with 12% DMSO was lower than in 15% DMSO, 4.4 and 3.0%, respectively. Formulation pH 9.0 buffer vs. pH 7.0 buffer did not have any effect on aggregation or yield, provided 10% sucrose was included in formulation. The percent aggregate in the 10%, 8%, 6%, and 4% (v/v) DMSO reactions were 2.7, 1.7, 1.0 and 0.5%, respectively. This suggests that CROl 1 and CROl IAE were very susceptible to aggregation when a higher percentage of DMSO is present.
AU four conjugation reactions resulted in a final molar ratio of 4.0 drugs/ Ab, suggesting that all four reactions went to completion. Safety margins for DMSO percentage in the conjugation reaction are 4 - 6%. This predicted to yield an aggregation level of 1% or less.
5. Investigation of Side Reaction During Conj ligation of CRO 11 to vcMMAE
Experiments were performed to: (1) investigate the extent and the kinetics of the side reaction in which maleimide-dmg is converted into an unreactive side product, which results in an incomplete conjugation and low drug-loading; (2) determine factors that influence the side reaction; and (3) determine whether the old vcMMAE lot (SGD 1006-0- 04) differed in reactivity compared to the new lot (SGDl 006-0-06).
Reactions (100 μl) containing vcMMAE at 30 μM final concentration, were incubated in borate pH 9.0 buffer either in the absence or in the presence of 2-fold molar excess of TCEP (with respect to vcMMAE). The reactions were quenched at 0, 2, 7 or 15 min with excess NAcCys. The control consisted of vcMMAE in phosphate pH 7.0 buffer quenched at the 15 min time point. The chromatograms are shown on Figure 23.
In pH 7 phosphate buffer 15 min and in pH 9.0 borate buffer 0 min after addition of the drug a single Cys-quenched product with (rt = 9.0 min) is formed (Compare A and B). In borate buffer pH 9.0 an unreactive side product is formed (rt=9.2 min) in a time- dependent fashion (B, C, D and E). In borate buffer and in the presence of TCEP (such as CROl 1 conjugation conditions), formation of the unreactive product is catalyzed resulting in >90% conversion of maleimide into succinimide after only 2 min of incubation (F through I). Both the old vcMMAE lot (SGD-1006-0-06) and the new lot (SGD-1006-0-04) exhibited similar reactivity towards high pH and TCEP, as well as similar kinetics.
Figure 24 shows the LC-MS identification of the unreactive product as succinimidyl- VCMMAE (rt = 9.2 min, m/z = 1318). Figure 25 shows the relative kinetics of formation of the succinimide in the presence or absence of TCEP.
Conclusions The side product is a result of conjugation performed at pH 9 instead of pH 7.4
(PBS). Formation of the side product is greatly enhanced in the presence of TCEP. The major stable side product has been identified by LC-MS as succinimidyl-vcMMAE. Minor and less stable side-products remain to be identified. Both vcMMAE lots behaved similarly. 6. Overcoming the Side Reaction During Conjugation of CROl 1 to vcMMAE
Experiments were performed to investigate whether the side reaction can be overcome by providing a larger excess of the drug for conjugation.
Several ways to suppress the side reaction were proposed: (1) Conducting conjugation at lower pH, e.g. 8.5 instead of 9.0 (high risk due to reduced solubility of CROl 1); (2) Removal of the excess of TCEP by UF/DF (not practical); and (3) Elevation of the excess of VCMMAE added upfront (practical).
100 mg of CROl 1 that was previously buffer exchanged into 50 mM borate-50 mM NaCl, was reduced with TCEP to generate 4.35 free thiols per Ab. The reaction was divided into two halves. For the first 50 mg half, a 10% excess of VCMMAE was added based on the 1 drug per thiol ratio . For the second half, a 20% excess was used. The conjugates were purified by UF/DF into 10 mM Histidine pH 6/ 10% sucrose solution. The results are summarized in Table 60.
Table 60. Preparation of CROl 1 -VCMMAE conjugates using various excess of vcMMAE based on the 1 drug per thiol ratio. Drug-to-Ab ratios were determined by RP HPLC.
VCMMAE Excess, %
10 20
SH per Ab 4.35 4.35 Drug-to-Ab ratio (in reaction) 3.9 4.1 Drug-to-Ab ratio (in final 3.7 4.0 product)
Conclusions
Using 10% versus 20% excess of vcMMAE was compared in a 100 mg conjugation. The higher excess of vcMMAE afforded a drug-to-Ab ratio closer to the expected value, and therefore, has been determined to be optimal.
Equivalents
The foregoing description and Examples detail certain preferred embodiments of the antibodies and describes the best mode contemplated by the inventors. It will be appreciated, however, that no matter how detailed the foregoing may appear in text, the methods of making and using the antibodies described herein may be practiced in many ways and the invention should be construed in accordance with the appended claims and any equivalents thereof. The foregoing written specification is considered to be sufficient to enable one skilled in the art to practice the embodiments described herein.

Claims

1. An isolated human antibody that specifically binds to GPNMB.
2. The antibody of claim 1 in which the antibody is a monoclonal antibody.
3. The antibody of claim 1 in which the antibody specifically binds GPNMB with an affinity constant greater than 107M"1.
4. The antibody of claim 1 comprising a region selected from the group comprising: VH1-2, VH2-5, VH3-11, VH3-21, VH3-30, VH3-33, VH4-31, VH4-59 and VH5-51.
5. The antibody of claim 4 further comprising a region selected from the group comprising: A2, A3, A20, A27, A30, L2 and 01.
6. The antibody of claim 1 comprising a region derived from a region selected from the group comprising: VHl -2, VH2-5, VH3-11, VH3-21, VH3-30, VH3-33, VH4-31, VH4-59 andVH5-51.
7. The antibody of claim 6 further comprising a region derived from a region selected from the group comprising: A2, A3, A20, A27, A30, L2 and 01.
8. The antibody of claim 1 comprising an amino acid sequence selected from the group comprising: SEQ ID NO: 2, 20, 38, 56, 74, 92, 110, 128, 146, 164, 182, 200, 218, 236, 253, 256, 260, 265, 270, 274, 277, 281 and 285.
9. The antibody of claim 8 further comprising an amino acid sequence selected from the group comprising: SEQ ID NO: 11, 29, 47, 65, 83, 101, 119, 137, 155, 173, 191,
209, 227 and 245.
10. The antibody of claim 1 comprising a CDR comprising an amino acid sequence selected from the group comprising: SEQ ID NO: 4, 6, 8, 13, 15, 17, 22, 24, 26, 31, 33, 35, 40, 42, 44, 49, 51, 53, 58, 60, 62, 67, 69, 71, 76, 78, 80, 85, 87, 89, 94, 96, 98, 103, 105, 107, 112, 114, 116, 121, 123, 125, 130, 132, 134, 139, 141, 143, 148,
150, 152, 157, 159, 161, 166, 168, 170, 175, 177, 179, 184, 186, 188, 193, 195, 197, 202, 204, 206, 211, 213, 215, 220, 222, 224, 229, 231, 233, 238, 240, 242, 247, 249, 251, 254, 257, 261, 266, 271, 278, 282, 286, 255, 258, 262, 267, 272, 275, 279, 283, 287, 259, 263, 264, 268, 269, 273, 276, 280, 284 and 288.
11. The antibody of claim 1 wherein the antibody is selected from the group comprising:
Mabl.10.2, Mabl.15.1, Mabl.2.2, Mabl.7.1, Mab2.10.2, Mab2.15.1, Mab2.16.1, Mab2.17.1, Mab2.21.2, Mab2.22.1, Mab2.24.1, Mab2.3.1, Mab2.7.1 and Mab2.8.1.
12. The antibody of any of the claims 1-11, wherein said antibody is an IgGl antibody.
13. An immunoconjugate comprising the antibody of claim 1 and a cytotoxic agent.
14. The immunoconjugate of claim 13, wherein said antibody is Mabl.15.1.
15. The immunoconjugate of claim 13, wherein the cytotoxic agent is auristatin E (dolastatin-10) or a derivative thereof.
16. A pharmaceutical composition comprising the immunoconjugate of claim 13.
17. A pharmaceutical composition comprising the antibody of claim 12, and an immunomodulator.
18. An antibody comprising human framework regions and means for specific binding to GPNMB.
19. The antibody of claim 18, wherein the means comprises a CDR comprising or derived from SEQ ID NO: 4, 6, 8, 13, 15, 17, 22, 24, 26, 31 , 33, 35, 40, 42, 44, 49, 51, 53, 58, 60, 62, 67, 69, 71, 76, 78, 80, 85, 87, 89, 94, 96, 98, 103, 105, 107, 112, 114, 116, 121, 123, 125, 130, 132, 134, 139, 141, 143, 148, 150, 152, 157, 159, 161, 166, 168, 170, 175, 177, 179, 184, 186, 188, 193, 195, 197, 202, 204, 206, 211, 213, 215, 220, 222, 224, 229, 231, 233, 238, 240, 242, 247, 249, 251, 254, 257, 261 ,
266, 271, 278, 282, 286, 255, 258, 262, 267, 272, 275, 279, 283, 287, 259, 263, 264, 268, 269, 273, 276, 280, 284 and 288.
20. An isolated nucleic acid encoding the antibody of claim 1.
21. An expression vector comprising the nucleic acid of claim 20.
22. A host cell comprising the vector of claim 21.
23. The host cell of claim 22, wherein the host cell is an E. coli bacterium, a Chinese hamster ovary cell, a HeLa cell, or a NSO cell.
24. The nucleic acid of claim 23, wherein the nucleic acid encodes the amino acid sequence selected from the group comprising: SEQ ID NO: 2, 20, 38, 56, 74, 92, 110, 128, 146, 164, 182, 200, 218, 236, 253, 256, 260, 265, 270, 274, 277, 281 and
285.
25. The nucleic acid of claim 24, wherein the nucleic acid comprises a nucleotide sequence selected from the group consisting of SEQ ID NO: 1, 19, 37, 55, 73, 91, 109, 127, 145, 163, 181, 199, 217, 235, 10, 28, 46, 64, 82, 100, 118, 136, 154, 172, 190, 208, 226 and 244.
26. A method of making a human monoclonal antibody that specifically binds with GPNMB, the method comprising the steps of: (a) providing a starting repertoire of nucleic acids encoding a variable domain that either includes a CDR3 to be replaced or lacks a CDR3 encoding region; (b) combining the repertoire with a donor nucleic acid encoding an amino acid sequence substantially as set out in the sequence selected from the group comprising: SEQ ID NO: 8, 26, 44, 62, 80, 98, 116, 134, 152, 170, 188, 206, 224, 242, 259, 263, 264, 268, 269, 273, 276, 280, 284 and 288 such that the donor nucleic acid is inserted into the CDR3 region in the repertoire, so as to provide a product repertoire of nucleic acids encoding a variable domain; (c) expressing the nucleic acids of the product repertoire; (d) selecting an antigen- binding fragment specific for GPNMB; and (e) recovering the specific antigen- binding fragment or nucleic acid encoding the binding fragment.
27. An antibody produced by the method of claim 26.
28. An antibody, or binding fragment thereof, that binds to GPNMB, wherein said antibody, or binding fragment thereof, neutralizes a GPNMB-induced activity, and wherein said antibody, or binding fragment thereof, cross-reacts with a fully human anti-GPNMB antibody selected from the group consisting of Mabl.2.1, MaM.10.1, and Mab2.22.1 or an antibody in the same antigen-binding bin as fully human anti-
GPNMB antibody Mabl .2.1, Mabl.10.1, or Mab2.22.1.
29. An antibody, or binding fragment thereof, that binds to GPNMB, wherein said antibody, or binding fragment thereof, neutralizes a GPNMB-induced activity, and wherein said antibody, or binding fragment thereof, cross-reacts with a fully human anti-GPNMB antibody selected from the group consisting of Mab2.3.1 and
Mabl.15.1 or an antibody in the same antigen-binding bin as fully human anti- GPNMB antibody Mab2.3.1 or Mab 1.15.1.
30. An antibody, or binding fragment thereof, that binds to GPNMB, wherein said antibody, or binding fragment thereof, neutralizes a GPNMB-induced activity, and wherein said antibody, or binding fragment thereof, cross-reacts with fully human anti-GPNMB antibody Mab2.10.1 or an antibody in the same antigen-binding bin as fully human anti-GPNMB antibody Mab2.10.1.
31. A single chain Fv antibody comprising a VL domain of a monoclonal human anti- GPNMB antibody linked to a VH domain of said anti-GPNMB antibody.
32. The single chain Fv antibody of claim 31 further comprising a VH domain of an anti-
CD3 antibody linked to a VL domain of said anti-CD3 antibody.
33. An immunoconjugate comprising a single clain Fv antibody of claim 31 and a cytotoxic agent.
34. The single chain Fv antibody of claim 31 or 32 further comprising a signal peptide or a flag tag.
35. The single chain Fv antibody of claim 31 comprising the amino acid sequence of SEQ ID NO: 355.
36. The single chain Fv antibody of claim 32 comprising the amino acid sequence of
SEQ ID NO: 357 or SEQ ID NO: 359.
37. A method of treating or preventing a desease associated with overexpression of GPNMB comprising admistering to a subject in need thereof an effective amount of a composition comprising (a) the antibody of claim 12;
(b) the immunoconjugate of claim 13 or 33; or
(c) the single chain Fv antibody of claim 32.
38. The method of claim 37, wherein said disease is melanoma or a neoplasm of CNS system.
39. The method of claim 38, wherein said neoplasm of CNS system is astrocytoma, glioblastoma, medulloblastoma, or neoplastic meningitis.
40. The method of claim 37, wherein said subject is human.
41. The method of claim 37, wherein said method comprises administering to said subject the immunoconjugate of claim 12, and wherein said effective amount is a unit dose between 0.1 mg/kg to 10 mg/kg, with 2 to 4 administrations.
42. The method of claim 41, wherein said effective amount is a unit dose between 0.1 mg/kg to 2 mg/kg.
43. The method of claim 41, wherein said effective amount is a unit dose about 1 mg/kg.
44. A method of detecting GPNMB-overexpressed cells in a biology sample comprising contacting said sample with a composition comprising the antibody of any of claims
1-11.
45. The method of claim 44, wherein said method is immunohistochemistry.
PCT/US2005/043482 2004-11-30 2005-11-30 Antibodies directed to gpnmb and uses thereof WO2006071441A2 (en)

Priority Applications (10)

Application Number Priority Date Filing Date Title
DE602005022928T DE602005022928D1 (en) 2004-11-30 2005-11-30 ANTIBODIES AGAINST GPNMB AND ITS USES
AT05852645T ATE476994T1 (en) 2004-11-30 2005-11-30 ANTIBODIES TO GPNMB AND THEIR USES
DK05852645.0T DK1827492T3 (en) 2004-11-30 2005-11-30 Antibodies targeting GPNMB and uses thereof
CA2589374A CA2589374C (en) 2004-11-30 2005-11-30 Antibodies directed to gpnmb and uses thereof
AU2005322410A AU2005322410B2 (en) 2004-11-30 2005-11-30 Antibodies directed to GPNMB and uses thereof
EP05852645A EP1827492B1 (en) 2004-11-30 2005-11-30 Antibodies directed to gpnmb and uses thereof
JP2007543622A JP2008521411A (en) 2004-11-30 2005-11-30 Antibodies against GPNMB and uses thereof
US13/355,366 US8846873B2 (en) 2004-11-30 2012-01-20 Antibodies directed to GPNMB and uses thereof
US14/454,179 US20150044212A1 (en) 2004-11-30 2014-08-07 Antibodies Directed to GPNMB and Uses Thereof
US15/053,204 US20160311919A1 (en) 2004-11-30 2016-02-25 Antibodies Directed To GPNMB And Uses Thereof

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US63202304P 2004-11-30 2004-11-30
US60/632,023 2004-11-30
US73377905P 2005-11-07 2005-11-07
US60/733,779 2005-11-07

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US11792032 A-371-Of-International 2005-11-30
US29077908A Continuation 2004-11-30 2008-11-03

Publications (2)

Publication Number Publication Date
WO2006071441A2 true WO2006071441A2 (en) 2006-07-06
WO2006071441A3 WO2006071441A3 (en) 2007-02-22

Family

ID=36615369

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2005/043482 WO2006071441A2 (en) 2004-11-30 2005-11-30 Antibodies directed to gpnmb and uses thereof

Country Status (9)

Country Link
US (3) US8846873B2 (en)
EP (3) EP2842571A1 (en)
JP (5) JP2008521411A (en)
AT (1) ATE476994T1 (en)
AU (1) AU2005322410B2 (en)
CA (1) CA2589374C (en)
DE (1) DE602005022928D1 (en)
DK (1) DK1827492T3 (en)
WO (1) WO2006071441A2 (en)

Cited By (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007053718A1 (en) * 2005-10-31 2007-05-10 The Government Of The United States, As Represented By The Secretary Of Health And Human Services, National Institutes Of Health Antibodies and immunotoxins that target human glycoprotein nmb
WO2008133641A2 (en) * 2006-10-11 2008-11-06 Curagen Corporation Antibodies directed to gpnmb and uses thereof
US7745394B2 (en) 2003-11-06 2010-06-29 Seattle Genetics, Inc. Monomethylvaline compounds capable of conjugation to ligands
US7829531B2 (en) 2002-07-31 2010-11-09 Seattle Genetics Inc. Drug conjugates and their use for treating cancer, an autoimmune disease or an infectious disease
WO2010135547A1 (en) * 2009-05-20 2010-11-25 Celldex Therapeutics, Inc. Antibodies directed to gpnmb and uses thereof
WO2012143495A3 (en) * 2011-04-21 2012-12-13 Bayer Intellectual Property Gmbh Novel binder-drug conjugates (adcs) and their use
US8460884B2 (en) 2008-11-05 2013-06-11 Los Angeles Biomedical Research Institute At Harbor-Ucla Medical Center Use of hematopoietic growth factor inducible neurokinin-1 (HGFIN) as a biomarker for renal injury or renal disease
US20130156784A1 (en) * 2008-08-20 2013-06-20 Michael Jeffers Compositions Using Antibodies Directed To GPNMB And Uses Thereof
WO2014114800A1 (en) * 2013-01-25 2014-07-31 Amgen Research (Munich) Gmbh Antibody constructs for cdh19 and cd3
WO2014114801A1 (en) * 2013-01-25 2014-07-31 Amgen Inc. Antibodies targeting cdh19 for melanoma
CN104914247A (en) * 2009-11-07 2015-09-16 阿斯图特医药公司 Method and composition for diagnosis and prognosis of renal injury and renal failure
WO2016016412A1 (en) * 2014-07-31 2016-02-04 Amgen Research (Munich) Gmbh Antibody constructs for cdh19 and cd3
WO2016016415A1 (en) * 2014-07-31 2016-02-04 Amgen Research (Munich) Gmbh Bispecific single chain antibody construct with enhanced tissue distribution
EP3003387A1 (en) * 2013-06-04 2016-04-13 Cytomx Therapeutics Inc. Compositions and methods for conjugating activatable antibodies
US9718883B2 (en) 2003-09-10 2017-08-01 Amgen Fremont Inc. Antibodies to M-CSF
US10011661B2 (en) * 2014-02-12 2018-07-03 Genentech, Inc. Anti-Jagged1 antibodies and methods of use
US10494432B2 (en) 2007-07-16 2019-12-03 Genentech, Inc. Anti-CD79B antibodies and immunoconjugates and methods of use
US10544218B2 (en) 2008-01-31 2020-01-28 Genentech, Inc. Anti-CD79B antibodies and immunoconjugates and methods of use
WO2020031936A1 (en) 2018-08-06 2020-02-13 第一三共株式会社 Combination of antibody-drug conjugate and tubulin inhibitor
US10981987B2 (en) 2007-07-16 2021-04-20 Genentech, Inc. Humanized anti-CD79b antibodies and immunoconjugates and methods of use
US11000510B2 (en) 2014-09-23 2021-05-11 Genentech, Inc. Methods of using anti-CD79b immunoconjugates
US11002738B2 (en) 2015-09-14 2021-05-11 Ventana Medical Systems, Inc. Anti-GPNMB antibodies and diagnostic uses thereof
WO2022212836A1 (en) * 2021-04-01 2022-10-06 Pyxis Oncology, Inc. Gpnmb antibodies and methods of use
WO2024026447A1 (en) 2022-07-29 2024-02-01 Alector Llc Anti-gpnmb antibodies and methods of use thereof

Families Citing this family (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DK1827492T3 (en) * 2004-11-30 2010-11-22 Curagen Corp Antibodies targeting GPNMB and uses thereof
HUE045435T2 (en) 2012-10-12 2019-12-30 Medimmune Ltd Pyrrolobenzodiazepines and conjugates thereof
WO2014140174A1 (en) 2013-03-13 2014-09-18 Spirogen Sàrl Pyrrolobenzodiazepines and conjugates thereof
JP6531166B2 (en) 2014-09-10 2019-06-12 メドイミューン・リミテッドMedImmune Limited Pyrrolobenzodiazepine and its conjugate
WO2016145022A1 (en) * 2015-03-11 2016-09-15 The Board Of Regents Of The University Of Texas System Anti-dc-hil antibodies for cancer diagnosis, prognosis and therapy
US20200023072A1 (en) 2016-10-11 2020-01-23 Medimmune Limited Antibody-drug conjugates with immune-mediated therapy agents
WO2019224275A1 (en) 2018-05-23 2019-11-28 Adc Therapeutics Sa Molecular adjuvant
JP6484385B1 (en) * 2018-11-28 2019-03-13 シーピー化成株式会社 Packaging container
US20220175950A1 (en) * 2018-12-03 2022-06-09 Agensys, Inc. Pharmaceutical compositions comprising anti-191p4d12 antibody drug conjugates and methods of use thereof
PE20212185A1 (en) 2019-02-18 2021-11-11 Lilly Co Eli FORMULATION OF THERAPEUTIC ANTIBODIES
KR102239781B1 (en) * 2019-04-08 2021-04-13 주식회사 녹십자 Bispecific antibody that specifically binds to gpnmb and cd3 and use thereof
KR20230031981A (en) 2019-05-14 2023-03-07 프로벤션 바이오, 인코포레이티드 Methods and compositions for preventing type 1 diabetes
US20230372528A1 (en) 2020-10-16 2023-11-23 University Of Georgia Research Foundation, Inc. Glycoconjugates
GB202102396D0 (en) 2021-02-19 2021-04-07 Adc Therapeutics Sa Molecular adjuvant

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4179337A (en) 1973-07-20 1979-12-18 Davis Frank F Non-immunogenic polypeptides
US4301144A (en) 1979-07-11 1981-11-17 Ajinomoto Company, Incorporated Blood substitute containing modified hemoglobin
US4496689A (en) 1983-12-27 1985-01-29 Miles Laboratories, Inc. Covalently attached complex of alpha-1-proteinase inhibitor with a water soluble polymer
US4640835A (en) 1981-10-30 1987-02-03 Nippon Chemiphar Company, Ltd. Plasminogen activator derivatives
US4670417A (en) 1985-06-19 1987-06-02 Ajinomoto Co., Inc. Hemoglobin combined with a poly(alkylene oxide)
US4791192A (en) 1986-06-26 1988-12-13 Takeda Chemical Industries, Ltd. Chemically modified protein with polyethyleneglycol

Family Cites Families (88)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS5896026A (en) 1981-10-30 1983-06-07 Nippon Chemiphar Co Ltd Novel urokinase derivative, its preparation and thrombolytic agent containing the same
US4609546A (en) 1982-06-24 1986-09-02 Japan Chemical Research Co., Ltd. Long-acting composition
US4522811A (en) 1982-07-08 1985-06-11 Syntex (U.S.A.) Inc. Serial injection of muramyldipeptides and liposomes enhances the anti-infective activity of muramyldipeptides
US4486414A (en) 1983-03-21 1984-12-04 Arizona Board Of Reagents Dolastatins A and B cell growth inhibitory substances
US4681581A (en) 1983-12-05 1987-07-21 Coates Fredrica V Adjustable size diaper and folding method therefor
US4766106A (en) 1985-06-26 1988-08-23 Cetus Corporation Solubilization of proteins for pharmaceutical compositions using polymer conjugation
US4735210A (en) 1985-07-05 1988-04-05 Immunomedics, Inc. Lymphographic and organ imaging method and kit
US5101827A (en) 1985-07-05 1992-04-07 Immunomedics, Inc. Lymphographic and organ imaging method and kit
US5776093A (en) 1985-07-05 1998-07-07 Immunomedics, Inc. Method for imaging and treating organs and tissues
AU597574B2 (en) 1986-03-07 1990-06-07 Massachusetts Institute Of Technology Method for enhancing glycoprotein stability
US4880935A (en) 1986-07-11 1989-11-14 Icrf (Patents) Limited Heterobifunctional linking agents derived from N-succinimido-dithio-alpha methyl-methylene-benzoates
US4831175A (en) 1986-09-05 1989-05-16 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Backbone polysubstituted chelates for forming a metal chelate-protein conjugate
US5099069A (en) 1986-09-05 1992-03-24 Gansow Otto A Backbone polysubstituted chelates for forming a metal chelate-protein conjugate
US5246692A (en) 1986-09-05 1993-09-21 The United States Of America As Represented By The Secretary Of Health And Human Services Backbone polysubstituted chelates for forming a metal chelate-protein conjugate
EP0307434B2 (en) 1987-03-18 1998-07-29 Scotgen Biopharmaceuticals, Inc. Altered antibodies
US4816444A (en) 1987-07-10 1989-03-28 Arizona Board Of Regents, Arizona State University Cell growth inhibitory substance
US5648471A (en) 1987-12-03 1997-07-15 Centocor, Inc. One vial method for labeling antibodies with Technetium-99m
IL106992A (en) 1988-02-11 1994-06-24 Bristol Myers Squibb Co Acylhydrazone derivatives of anthracycline and methods for their preparation
GB8823869D0 (en) 1988-10-12 1988-11-16 Medical Res Council Production of antibodies
US5076973A (en) 1988-10-24 1991-12-31 Arizona Board Of Regents Synthesis of dolastatin 3
US5175384A (en) 1988-12-05 1992-12-29 Genpharm International Transgenic mice depleted in mature t-cells and methods for making transgenic mice
US4978744A (en) 1989-01-27 1990-12-18 Arizona Board Of Regents Synthesis of dolastatin 10
US4879278A (en) 1989-05-16 1989-11-07 Arizona Board Of Regents Isolation and structural elucidation of the cytostatic linear depsipeptide dolastatin 15
US4986988A (en) 1989-05-18 1991-01-22 Arizona Board Of Regents Isolation and structural elucidation of the cytostatic linear depsipeptides dolastatin 13 and dehydrodolastatin 13
EP0486622B1 (en) 1989-08-09 1998-11-04 Rhomed, Incorporated Direct radiolabeling of antibodies and other proteins with technetium or rhenium
US5460785A (en) 1989-08-09 1995-10-24 Rhomed Incorporated Direct labeling of antibodies and other protein with metal ions
US5138036A (en) 1989-11-13 1992-08-11 Arizona Board Of Regents Acting On Behalf Of Arizona State University Isolation and structural elucidation of the cytostatic cyclodepsipeptide dolastatin 14
ATE139258T1 (en) 1990-01-12 1996-06-15 Cell Genesys Inc GENERATION OF XENOGENE ANTIBODIES
US6150584A (en) 1990-01-12 2000-11-21 Abgenix, Inc. Human antibodies derived from immunized xenomice
US6075181A (en) 1990-01-12 2000-06-13 Abgenix, Inc. Human antibodies derived from immunized xenomice
US6673986B1 (en) 1990-01-12 2004-01-06 Abgenix, Inc. Generation of xenogeneic antibodies
US5124471A (en) 1990-03-26 1992-06-23 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Bifunctional dtpa-type ligand
GB9015198D0 (en) 1990-07-10 1990-08-29 Brien Caroline J O Binding substance
US5877397A (en) 1990-08-29 1999-03-02 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US5661016A (en) 1990-08-29 1997-08-26 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US5814318A (en) 1990-08-29 1998-09-29 Genpharm International Inc. Transgenic non-human animals for producing heterologous antibodies
US5874299A (en) 1990-08-29 1999-02-23 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5612205A (en) 1990-08-29 1997-03-18 Genpharm International, Incorporated Homologous recombination in mammalian cells
US6255458B1 (en) 1990-08-29 2001-07-03 Genpharm International High affinity human antibodies and human antibodies against digoxin
US5633425A (en) 1990-08-29 1997-05-27 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5789650A (en) 1990-08-29 1998-08-04 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
US5625126A (en) 1990-08-29 1997-04-29 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
US5770429A (en) 1990-08-29 1998-06-23 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5545806A (en) 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
US5194594A (en) 1990-09-07 1993-03-16 Techniclone, Inc. Modified antibodies
AU2515992A (en) 1991-08-20 1993-03-16 Genpharm International, Inc. Gene targeting in animal cells using isogenic dna constructs
US5565332A (en) 1991-09-23 1996-10-15 Medical Research Council Production of chimeric antibodies - a combinatorial approach
US5622929A (en) 1992-01-23 1997-04-22 Bristol-Myers Squibb Company Thioether conjugates
US6034065A (en) 1992-12-03 2000-03-07 Arizona Board Of Regents Elucidation and synthesis of antineoplastic tetrapeptide phenethylamides of dolastatin 10
US5635483A (en) 1992-12-03 1997-06-03 Arizona Board Of Regents Acting On Behalf Of Arizona State University Tumor inhibiting tetrapeptide bearing modified phenethyl amides
US5981175A (en) 1993-01-07 1999-11-09 Genpharm Internation, Inc. Methods for producing recombinant mammalian cells harboring a yeast artificial chromosome
US5410024A (en) 1993-01-21 1995-04-25 Arizona Board Of Regents Acting On Behalf Of Arizona State University Human cancer inhibitory pentapeptide amides
US5780588A (en) 1993-01-26 1998-07-14 Arizona Board Of Regents Elucidation and synthesis of selected pentapeptides
US6214345B1 (en) 1993-05-14 2001-04-10 Bristol-Myers Squibb Co. Lysosomal enzyme-cleavable antitumor drug conjugates
US5625825A (en) 1993-10-21 1997-04-29 Lsi Logic Corporation Random number generating apparatus for an interface unit of a carrier sense with multiple access and collision detect (CSMA/CD) ethernet data network
US5530097A (en) 1994-08-01 1996-06-25 Arizona Board Of Regents Acting On Behalf Of Arizona State University Human cancer inhibitory peptide amides
US5521284A (en) 1994-08-01 1996-05-28 Arizona Board Of Regents Acting On Behalf Of Arizona State University Human cancer inhibitory pentapeptide amides and esters
US5504191A (en) 1994-08-01 1996-04-02 Arizona Board Of Regents Acting On Behalf Of Arizona State University Human cancer inhibitory pentapeptide methyl esters
US5554725A (en) 1994-09-14 1996-09-10 Arizona Board Of Regents Acting On Behalf Of Arizona State University Synthesis of dolastatin 15
US5643763A (en) 1994-11-04 1997-07-01 Genpharm International, Inc. Method for making recombinant yeast artificial chromosomes by minimizing diploid doubling during mating
US5599902A (en) 1994-11-10 1997-02-04 Arizona Board Of Regents Acting On Behalf Of Arizona State University Cancer inhibitory peptides
US5663149A (en) 1994-12-13 1997-09-02 Arizona Board Of Regents Acting On Behalf Of Arizona State University Human cancer inhibitory pentapeptide heterocyclic and halophenyl amides
US6030613A (en) 1995-01-17 2000-02-29 The Brigham And Women's Hospital, Inc. Receptor specific transepithelial transport of therapeutics
DE69626849T2 (en) 1995-12-22 2003-12-24 Bristol Myers Squibb Co BRANCHED HYDRAZONE GROUPS OF COUPLERS
WO1998036765A1 (en) 1997-02-25 1998-08-27 Arizona Board Of Regents Isolation and structural elucidation of the cytostatic linear and cyclo-depsipeptides dolastatin 16, dolastatin 17, and dolastatin 18
US6960570B2 (en) 1998-03-18 2005-11-01 Corixa Corporation Compositions and methods for the therapy and diagnosis of lung cancer
US20030100720A1 (en) 1998-11-18 2003-05-29 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US6323315B1 (en) 1999-09-10 2001-11-27 Basf Aktiengesellschaft Dolastatin peptides
US6812002B2 (en) * 2000-08-30 2004-11-02 Pfizer Inc. Osteoactivin protein and nucleic acids encoding the same, compositions and methods of stimulating bone differentiation
US20020102264A1 (en) * 2000-10-18 2002-08-01 Cheung Nai-Kong V. Uses of monoclonal antibody 8H9
WO2002062947A2 (en) 2000-10-20 2002-08-15 University Of Medicine And Dentistry New Jersey Medical School Hematopoietic growth factor inducible neurokinin-1 gene
US20030202938A1 (en) * 2000-10-20 2003-10-30 Pranela Rameshwar Hematopoietic growth factor inducible neurokinin-1 gene
WO2002046475A2 (en) 2000-12-08 2002-06-13 Curagen Corporation Method of detecting and treating tuberous sclerosis complex associated disorders
US6884869B2 (en) 2001-04-30 2005-04-26 Seattle Genetics, Inc. Pentapeptide compounds and uses related thereto
US20030083263A1 (en) 2001-04-30 2003-05-01 Svetlana Doronina Pentapeptide compounds and uses related thereto
US7115265B1 (en) 2001-05-14 2006-10-03 Duke University Four genetic tumor markers specific for human glioblastoma
US6441163B1 (en) 2001-05-31 2002-08-27 Immunogen, Inc. Methods for preparation of cytotoxic conjugates of maytansinoids and cell binding agents
WO2003048731A2 (en) 2001-12-03 2003-06-12 Abgenix, Inc. Antibody categorization based on binding characteristics
AU2003228336B2 (en) 2002-03-19 2009-07-30 Curagen Corporation Therapeutic polypeptides, nucleic acids encoding same, and methods of use
PT1545613E (en) 2002-07-31 2011-09-27 Seattle Genetics Inc Auristatin conjugates and their use for treating cancer, an autoimmune disease or an infectious disease
EP2289559B1 (en) 2003-02-20 2014-02-12 Seattle Genetics, Inc. Anit-CD70 antibody-drug conjugates and their use for the treatment of cancer and immune disorders
AU2004224390A1 (en) * 2003-03-19 2004-10-07 Abgenix, Inc. Antibodies against T cell immunoglobulin domain and mucin domain 1 (TIM-1) antigen and uses thereof
KR101531400B1 (en) * 2003-06-27 2015-06-26 암젠 프레몬트 인코포레이티드 Antibodies directed to the deletion mutants of epidermal growth factor receptor and uses thereof
US20050026881A1 (en) * 2003-07-31 2005-02-03 Robinson Cynthia B. Combination of dehydroepiandrosterone or dehydroepiandrosterone-sulfate with an anti-IgE antibody for treatment of asthma or chronic obstructive pulmonary disease
US7968684B2 (en) * 2003-11-12 2011-06-28 Abbott Laboratories IL-18 binding proteins
DK1827492T3 (en) * 2004-11-30 2010-11-22 Curagen Corp Antibodies targeting GPNMB and uses thereof
US20130156784A1 (en) * 2008-08-20 2013-06-20 Michael Jeffers Compositions Using Antibodies Directed To GPNMB And Uses Thereof
EP2453919A1 (en) * 2009-05-20 2012-05-23 Celldex Therapeutics, Inc. Antibodies directed to gpnmb and uses thereof

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4179337A (en) 1973-07-20 1979-12-18 Davis Frank F Non-immunogenic polypeptides
US4301144A (en) 1979-07-11 1981-11-17 Ajinomoto Company, Incorporated Blood substitute containing modified hemoglobin
US4640835A (en) 1981-10-30 1987-02-03 Nippon Chemiphar Company, Ltd. Plasminogen activator derivatives
US4496689A (en) 1983-12-27 1985-01-29 Miles Laboratories, Inc. Covalently attached complex of alpha-1-proteinase inhibitor with a water soluble polymer
US4670417A (en) 1985-06-19 1987-06-02 Ajinomoto Co., Inc. Hemoglobin combined with a poly(alkylene oxide)
US4791192A (en) 1986-06-26 1988-12-13 Takeda Chemical Industries, Ltd. Chemically modified protein with polyethyleneglycol

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
"Proteins, Structures and Molecular Principles", 1984, W. H. FREEMAN AND COMPANY

Cited By (62)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7829531B2 (en) 2002-07-31 2010-11-09 Seattle Genetics Inc. Drug conjugates and their use for treating cancer, an autoimmune disease or an infectious disease
US7851437B2 (en) 2002-07-31 2010-12-14 Seattle Genetics Inc. Drug conjugates and their use for treating cancer, an autoimmune disease or an infectious disease
US8906376B2 (en) 2002-07-31 2014-12-09 Seattle Genetics, Inc. Drug conjugates and their use for treating cancer, an autoimmune disease or an infectious disease
US10280219B2 (en) 2003-09-10 2019-05-07 Amgen Fremont Inc. Antibodies to M-CSF
US9718883B2 (en) 2003-09-10 2017-08-01 Amgen Fremont Inc. Antibodies to M-CSF
US8557780B2 (en) 2003-11-06 2013-10-15 Seattle Genetics, Inc. Monomethylvaline compounds capable of conjugation to ligands
US7745394B2 (en) 2003-11-06 2010-06-29 Seattle Genetics, Inc. Monomethylvaline compounds capable of conjugation to ligands
US10414826B2 (en) 2003-11-06 2019-09-17 Seattle Genetics, Inc. Monomethylvaline compounds capable of conjugation to ligands
US10808039B2 (en) 2003-11-06 2020-10-20 Seattle Genetics Inc. Monomethylvaline compounds capable of conjugation to ligands
US8703714B2 (en) 2003-11-06 2014-04-22 Seattle Genetics, Inc. Monomethylvaline compounds capable of conjugation to ligands
US8039593B2 (en) 2005-10-31 2011-10-18 Duke University Antibodies and immunotoxins that target human glycoprotein NMB
US9115196B2 (en) 2005-10-31 2015-08-25 The United States Of America As Represented By The Secretary Of Health Antibodies and immunotoxins that target human glycoprotein NMB
WO2007053718A1 (en) * 2005-10-31 2007-05-10 The Government Of The United States, As Represented By The Secretary Of Health And Human Services, National Institutes Of Health Antibodies and immunotoxins that target human glycoprotein nmb
US8445216B2 (en) 2005-10-31 2013-05-21 Duke University Antibodies and immunotoxins that target human glycoprotein NMB
US20130244255A1 (en) * 2006-10-11 2013-09-19 Vincent A. Pollack Antibodies directed to gpnmb and uses thereof
WO2008133641A2 (en) * 2006-10-11 2008-11-06 Curagen Corporation Antibodies directed to gpnmb and uses thereof
WO2008133641A3 (en) * 2006-10-11 2009-03-12 Curagen Corp Antibodies directed to gpnmb and uses thereof
US11866496B2 (en) 2007-07-16 2024-01-09 Genentech, Inc. Humanized anti-CD79B antibodies and immunoconjugates and methods of use
USRE48558E1 (en) 2007-07-16 2021-05-18 Genentech, Inc. Anti-CD79B antibodies and immunoconjugates and methods of use
US10494432B2 (en) 2007-07-16 2019-12-03 Genentech, Inc. Anti-CD79B antibodies and immunoconjugates and methods of use
US10981987B2 (en) 2007-07-16 2021-04-20 Genentech, Inc. Humanized anti-CD79b antibodies and immunoconjugates and methods of use
US10544218B2 (en) 2008-01-31 2020-01-28 Genentech, Inc. Anti-CD79B antibodies and immunoconjugates and methods of use
US20130156784A1 (en) * 2008-08-20 2013-06-20 Michael Jeffers Compositions Using Antibodies Directed To GPNMB And Uses Thereof
US8460884B2 (en) 2008-11-05 2013-06-11 Los Angeles Biomedical Research Institute At Harbor-Ucla Medical Center Use of hematopoietic growth factor inducible neurokinin-1 (HGFIN) as a biomarker for renal injury or renal disease
WO2010135547A1 (en) * 2009-05-20 2010-11-25 Celldex Therapeutics, Inc. Antibodies directed to gpnmb and uses thereof
US20130058948A1 (en) * 2009-05-20 2013-03-07 Michael Jeffers Antibodies Directed To Gpnmb And Uses Thereof
CN104914247A (en) * 2009-11-07 2015-09-16 阿斯图特医药公司 Method and composition for diagnosis and prognosis of renal injury and renal failure
EP3153863A1 (en) * 2009-11-07 2017-04-12 Astute Medical, Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure
US8992932B2 (en) 2011-04-21 2015-03-31 Seattle Genetics, Inc. Binder-drug conjugates (ADCs) and use thereof
RU2610336C2 (en) * 2011-04-21 2017-02-09 Сиэтл Дженетикс, Инк. New conjugates binding compound - active compound (adc) and use thereof
WO2012143495A3 (en) * 2011-04-21 2012-12-13 Bayer Intellectual Property Gmbh Novel binder-drug conjugates (adcs) and their use
US9920121B2 (en) 2013-01-25 2018-03-20 Amgen Inc. Antibodies targeting CDH19 for melanoma
IL275114A (en) * 2013-01-25 2020-07-30 Amgen Inc Antibody constructs for binding domains for cdh19 and cd3, compositions comprising same, methods of producing same and uses thereof
US10059766B2 (en) 2013-01-25 2018-08-28 Amgen Inc. Antibody constructs for CDH19 and CD3
US11053311B2 (en) 2013-01-25 2021-07-06 Amgen Inc. Antibodies targeting CDH19 for melanoma
TWI660971B (en) * 2013-01-25 2019-06-01 德商安美基研究(慕尼黑)公司 Antibody constructs for cdh19 and cd3
KR20150134322A (en) * 2013-01-25 2015-12-01 암젠 리서치 (뮌헨) 게엠베하 Antibody constructs for cdh19 and cd3
WO2014114800A1 (en) * 2013-01-25 2014-07-31 Amgen Research (Munich) Gmbh Antibody constructs for cdh19 and cd3
KR20210038682A (en) * 2013-01-25 2021-04-07 암젠 리서치 (뮌헨) 게엠베하 Antibody constructs for cdh19 and cd3
KR102230620B1 (en) * 2013-01-25 2021-03-23 암젠 리서치 (뮌헨) 게엠베하 Antibody constructs for cdh19 and cd3
US11498964B2 (en) 2013-01-25 2022-11-15 Amgen Research (Munich) Gmbh Antibody constructs for CDH19 and CD3
TWI695012B (en) * 2013-01-25 2020-06-01 德商安美基研究(慕尼黑)公司 Antibody constructs for cdh19 and cd3
WO2014114801A1 (en) * 2013-01-25 2014-07-31 Amgen Inc. Antibodies targeting cdh19 for melanoma
EP3699194A1 (en) * 2013-01-25 2020-08-26 Amgen Research (Munich) GmbH Antibody constructs for cdh19 and cd3
CN105121468A (en) * 2013-01-25 2015-12-02 安进研发(慕尼黑)股份有限公司 Antibody constructs for CDH19 and CD3
KR102452351B1 (en) 2013-01-25 2022-10-07 암젠 리서치 (뮌헨) 게엠베하 Antibody constructs for cdh19 and cd3
CN105121468B (en) * 2013-01-25 2020-11-10 安进研发(慕尼黑)股份有限公司 Antibody constructs for CDH19 and CD3
EA036950B1 (en) * 2013-01-25 2021-01-19 Эмджен Рисерч (Мюник) Гмбх Antibody constructs for cdh19 and cd3
AU2014274982B2 (en) * 2013-06-04 2019-12-05 Cytomx Therapeutics, Inc Compositions and methods for conjugating activatable antibodies
EP3003387A1 (en) * 2013-06-04 2016-04-13 Cytomx Therapeutics Inc. Compositions and methods for conjugating activatable antibodies
US10858440B2 (en) 2014-02-12 2020-12-08 Genentech, Inc. Anti-Jagged1 antibodies and methods of use
US10011661B2 (en) * 2014-02-12 2018-07-03 Genentech, Inc. Anti-Jagged1 antibodies and methods of use
US11926674B2 (en) 2014-02-12 2024-03-12 Genentech, Inc. Anti-Jagged2 antibodies and methods of use
WO2016016412A1 (en) * 2014-07-31 2016-02-04 Amgen Research (Munich) Gmbh Antibody constructs for cdh19 and cd3
AU2015295242B2 (en) * 2014-07-31 2020-10-22 Amgen Research (Munich) Gmbh Bispecific single chain antibody construct with enhanced tissue distribution
WO2016016415A1 (en) * 2014-07-31 2016-02-04 Amgen Research (Munich) Gmbh Bispecific single chain antibody construct with enhanced tissue distribution
US20170275373A1 (en) * 2014-07-31 2017-09-28 Amgen Research (Munich) Gmbh Bispecific single chain antibody construct with enhanced tissue distribution
US11000510B2 (en) 2014-09-23 2021-05-11 Genentech, Inc. Methods of using anti-CD79b immunoconjugates
US11002738B2 (en) 2015-09-14 2021-05-11 Ventana Medical Systems, Inc. Anti-GPNMB antibodies and diagnostic uses thereof
WO2020031936A1 (en) 2018-08-06 2020-02-13 第一三共株式会社 Combination of antibody-drug conjugate and tubulin inhibitor
WO2022212836A1 (en) * 2021-04-01 2022-10-06 Pyxis Oncology, Inc. Gpnmb antibodies and methods of use
WO2024026447A1 (en) 2022-07-29 2024-02-01 Alector Llc Anti-gpnmb antibodies and methods of use thereof

Also Published As

Publication number Publication date
JP5992386B2 (en) 2016-09-14
JP2012120544A (en) 2012-06-28
JP2017099415A (en) 2017-06-08
EP2305716A2 (en) 2011-04-06
JP2008521411A (en) 2008-06-26
EP2305716A3 (en) 2011-05-25
US20150044212A1 (en) 2015-02-12
JP6334496B2 (en) 2018-05-30
AU2005322410A1 (en) 2006-07-06
CA2589374A1 (en) 2006-07-06
WO2006071441A3 (en) 2007-02-22
DK1827492T3 (en) 2010-11-22
CA2589374C (en) 2016-05-03
EP2305716B1 (en) 2014-10-22
JP2016026210A (en) 2016-02-12
ATE476994T1 (en) 2010-08-15
EP1827492A2 (en) 2007-09-05
US8846873B2 (en) 2014-09-30
EP2842571A1 (en) 2015-03-04
JP2014003986A (en) 2014-01-16
DE602005022928D1 (en) 2010-09-23
JP5716151B2 (en) 2015-05-13
US20130022597A1 (en) 2013-01-24
US20160311919A1 (en) 2016-10-27
AU2005322410B2 (en) 2011-11-10
EP1827492B1 (en) 2010-08-11

Similar Documents

Publication Publication Date Title
EP2305716B1 (en) Antibodies directed to gpnmb and uses thereof
US20130244255A1 (en) Antibodies directed to gpnmb and uses thereof
JP2023099088A (en) Activatable anti-pdl1 antibodies, and methods of use thereof
US8101180B2 (en) Anti-IGF-IR and/or anti-insulin/IGF-I hybrid receptors antibodies and uses thereof
US7914784B2 (en) Anti-IGF-IR antibodies and uses thereof
US20180043014A1 (en) Compositions Using Antibodies Directed to GPNMB and Uses Thereof
WO2005058967A2 (en) Novel anti-insulin/igf-i hybrid receptor or anti-insulin/igf-i hybrid receptor and igf-ir antibodies and uses thereof
TW201132352A (en) Anti-GCC antibody molecules and related compositions and methods
US20080063639A1 (en) Method for the treatment of psoriasis comprising novel anti-IGF-IR antibodies
US20050142137A1 (en) Antibodies against secretoryleukocyte protease inhibitor
US9375488B2 (en) Compounds to fibroblast growth factor receptor-3 (FGFR3) and methods of treatment
ES2359426T3 (en) ANTIBODIES DIRECTED TO GPNMB AND ITS APPLICATIONS.
JP2022550434A (en) Antibody compositions containing Fc mutations and site-specific conjugation properties

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 2007543622

Country of ref document: JP

Ref document number: 2589374

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2005322410

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2005852645

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2005322410

Country of ref document: AU

Date of ref document: 20051130

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 2005322410

Country of ref document: AU

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWP Wipo information: published in national office

Ref document number: 2005852645

Country of ref document: EP